The Basics of Bioanalysis - PowerPoint PPT Presentation

About This Presentation
Title:

The Basics of Bioanalysis

Description:

The Basics of Bioanalysis - Method Development, Method Validation & Study Sample Analysis – PowerPoint PPT presentation

Number of Views:98
Slides: 25
Provided by: TarakBTP
Tags:

less

Transcript and Presenter's Notes

Title: The Basics of Bioanalysis


1
B.TARAK PRASADAssistant Manager-BioanalyticalACT
IMUS BIOSCIENCES PVT LTD
  • The Basics of Bioanalysis How Do We Develop
    and Validate Your Bioanalytical Method?

2
Introduction
  • Bioanalytics is an essential tool in drug
    discovery and development for determining the
    concentration of drugs and their metabolites as
    well as various pharmacodynamic biomarkers in
    biological fluids.
  • In these analyses, scientists use developed and
    validated methods to quantitatively detect
    analytes and metabolites within biological
    matrices such as blood, serum, plasma, urine,
    etc.
  • A critical component of any bioanalytical program
    includes bioanalytical method validation,
    ensuring quantitative results demonstrate
    accuracy, precision, selectivity, and stability
    so the accuracy of sample analysis results can be
    justified.

3
Table of Content
  • Overview Definition
  • Bioanalytical Method Development
  • Bioanalytical Method Validation
  • Sample Analysis
  • Reporting
  • Regulatory Guidelines

4
Overview Definition
  • Bioanalysis isnt merely running standard
    analyses for a biological sample. We must produce
    results which are quantitative and valid as per
    FDA guidance as these analyses constitute the
    foundational block towards drug approval. Due to
    the significance of these analyses, regulatory
    authorities generally audit these results for
    accuracy before approval of the drug.
  • Its imperative to have the support and expertise
    of veterans, such as the team at Actimus BioLab,
    when the complexities of bioanalysis get
    cumbersome.
  • Our scientists help navigate bioanalytical method
    validation following method development focused
    on the appropriate detection range for your
    assay, as well as offer insights on the
    requirements of FDA and other international
    regulatory authorities.
  • This article alludes how our team can help you
    with custom assay development that meets all the
    criteria outlined above.

5
schematic diagram of bioanalytical workflow
Background of analyte, available dosage forms
etc..
Literature Review
Physical state of analyte, Pka, solubility,
chemical properties and structure of analyte etc..
Physical chemical properties of analyte
On the basis of Cmax data dose of drug, range
of quantification is selected
Setting the range of quantification
On the basis of nature and chemical properties of
analyte, analytical instrument is selected
Selection of analytical instrument
Mass spectrophotometer
HPLC
Optimisation of mass parameters
Selection of mobile phase internal standard
Optimisation of chromatographic conditions
Experiment using final parameter of method e.g.
precision and accuracy batches , selectivity ,
sensitivity etc. as per requirement
Optimisation of Extraction procedure
Evalution of analyte recovery and stability
6
Bioanalytical Method Development
  • Steps involved in the Method Development
  • Method selection and information of Sample
  • Initial method conditions
  • Processing the analytical method in aqueous
    standards
  • Development and optimization of sample processing
    method
  • Checking the analytical method in biological
    matrix
  • Pre-validation

7
1.Method selection and information of Sample
  • Literature survey shall be conducted to have
    first hand information on drug profile and its
    pharmacokinetic properties.
  • Collection of physicochemical properties of the
    analytes and the related compounds are essential
    for the development of the analytical method.
  • Based on the drugs physicochemical properties
    such as molecular size, shape, structure,
    functional groups, polarity, partition
    coefficient, solubility, dissociation constant
    etc.,
  • choose the internal standard having comparable
    molecular structure and physicochemical
    properties with respect to the analytes. Same
    molecule with different isotopes like deuterium,
    C13 and N15 will be a better alternative for
    internal standards.

8
2. Selection of initial method conditions
  • Setting the initial method conditions include
    diluent selection based on the solubility of the
    drug, drug metabolites and internal standard and
    compatibility with analytical method.
  • The lowest concentration to be quantified shall
    be assessed using aqueous solutions during this
    phase.
  • Run time and resolution between the peaks should
    be taken care during this phase

9
3. Parameters to be optimized
  • 1.Mass parameters
  • 2.LC parameters
  • Mode of separation
  • Selection of stationary phase
  • Selection of mobile phase

10
Mass parameters
  • DP (declustering potential) -The declustering
    potential (DP) is a voltage applied to the
    orifice that helps to prevent the ions from
    clustering together.
  • EP (entrance potential) -The rods in Q0 do not
    act as mass filters but serve to guide and focus
    the ions into the mass spectrometer. It is here
    that the entrance potential (EP) is applied.
  • CE (collision energy) -The collision energy (CE)
    refers to the rate of acceleration as the ions
    enter quadrupole 2 (Q2.). The ions undergo a
    thermal interaction with the collision gas and
    fragment.
  • CXP (collision cell exit potential)-The Collision
    Cell Exit Potential (CXP) focuses and accelerates
    the ions out of Q2 and into Q3.
  • CAD (collision gas) -Collisionally Activated
    Dissociation (CAD) is the process of colliding
    precursor ions (parent ions) with a neutral gas
    to break the molecule into fragment ions. The
    neutral gas used during this demonstration is
    nitrogen.
  • IS (ionspray voltage)
  • TEM (temperature of ion source)
  • GS1 (nebulizing gas)
  • GS2 (drying gas)

11
LC parameters
  • Mode of separation
  • In reverse phase mode, the mobile phase is
    comparatively more polar than the stationary
    phase. For the separation of polar or moderately
    polar compounds, the suitable mode is reverse
    phase. The nature of the analyte is the primary
    factory in the selection of mode of separation.

12
Selection of stationary phase
  • Selection of the column is the first and the most
    important step in method development, because the
    column is the heart of separation process.
  • The appropriate choice of separation column
    includes different approaches
  • ? Column dimensions
  • ? Nature of packing material
  • ? Shape of the particles
  • ? Size of the particles
  • ? Surface area
  • ? Pore volume
  • ? End capping

13
  • The optimum length of the column required for a
    particular separation is dictated by the number
    of theoretical plates needed to give the desired
    resolution. If the column is too short, then the
    clearly the column will not have enough
    resolving power to achieve the separation and
    if it is too long, then analysis time is
    needlessely extended. The most common column
    lengths used in regular analytical HPLC are 10,
    12.5, 15 and 25 cm, with 15 cm columns being
    perhaps the most popular.
  • Currently, most HPLC separation are carried out
    with 5µm diameter packing materials. Columns with
    5µm particle size give the best compromise of
    efficiency, reproducibility and reliability.
  • As the particle size decreases the surface area
    for coating increases. Generally high specific
    surface area will increase the retention of
    solutes by increasing the capacity factor.
  • Reverse phase mode of chromatography facilitates
    a wide range of columns like dimethylsilane (C2),
    butylsilane (C4), octyl silane (C8), octadecyl
    silane (C18), cyanopropyl (CN), nitro, amino etc.

14
Selection of Mobile phase
  • The primary objective in selection and
    optimization of mobile phase is to achieve
    optimum separation of all the individual
    impurities and degradants from each other and
    analyte peak.
  • The following are the parameters to be considered
    during selection and optimization of mobile
    phase.
  • ? Buffer
  • ? pH of the buffer
  • ? Mobile phase composition

15
  • Buffer - Buffer and its strength play an
    important role in deciding the peak symmetries
    and separations. The retention time depends on
    molar strength of buffer. Molar strength is
    proportional to retention time. In order to
    achieve better separation the strength of the
    buffer can be increased.
  • pH of the buffer- pH plays an important role in
    achieving the chromatographic separation as it
    controls the elution properties by controlling
    the ionization characteristics. A different
    concentration of buffer was chosen to achieve
    required separations. It is important to maintain
    the pH of mobile phase in the range of 2.0 to 8.0
    as most of the columns does not withstand out of
    this range 12. As Siloxane linkages are cleaved
    below pH 2 and at above pH 8 silica dissolves.

16
4.Checking the analytical method in aqueous
standards
  • Before going to analyze a method in biological
    matrix, first check the analytical method in
    aqueous standards.
  • Prepare aqueous calibration curve standards, at
    least with four concentrations, including the
    highest and lowest. Concentration of the highest
    standard shall be based on Cmax and lowest
    standard shall be tentatively fixed based on the
    preliminary studies. Make injections of each
    calibration curve standard and find the
    correlation coefficient. Correlation co-efficient
    (r) should not be less than 0.99.
  • If required, adjust the mobile phase, mass
    spectral parameters (if applicable) and
    chromatographic conditions such as mobile phase
    constituents, buffer strength, ratio, pH, flow
    rate, column, column oven temperature etc., to
    get the clear resolution with required
    sensitivity.

17
5. Development and optimization of sample
processing method
  • When the instrumental method is concluded with
    aqueous standards, prepare matrix sample. Based
    on the literature survey data on analyte and
    internal standards physicochemical properties
    like structure, functional groups, pH, partition
    coefficient, dissociation constant, polarity and
    solubility, set and optimize the sample
    preparation technique like
  • protein precipitation
  • liquid-liquid extraction
  • solid phase extraction

18
(No Transcript)
19
Bioanalytical Method Validation
  • Method validation is a part of GLP study and it
    is to ensure the quality of analytical method or
    it is a process of demonstrating that analytical
    method is suitable for its intended use. Method
    validation is applied to ensure the method relia
  • A typical Full Bioanalytical Method Validation
    should include determination of
  • 1. System suitability 2. Selectivity 3.
    Sensitivity 4. Precision and Accuracy i)
    Inter-Day Precision and Accuracy ii) Intra-Day
    Precision and Accuracy 5. Recovery 6. Calibration
    Curve 7. Matrix effect 8. Haemolyzed effect 9.
    Lipemic effect 10. Dilution integrity 11.
    Stability 12. Auto sampler carry over 13.
    Reinjection reproducibility 14. Ruggednessbility,
    quality and reproducibility.
  • Acceptance Criteria of an Analytical Runs
  • For Blank sample and Blank IS sample
  • If any peak area is present at the
    retention time of analyte in Blank or Blank IS
    sample, its area response should be lt 20.00 of
    analyte area response of LLOQ standard. If any
    peak area is present at the retention time of
    Internal standard in Blank sample, its area
    response should be lt 5.00 of the IS area
    response of LLOQ standard.
  • At least 75 of calibrators must pass 15 of the
    nominal concentration (20 at the LLOQ)
  • RSD for LLOQ QC level Not More Than 20.00
  • RSD for samples other than LLOQ QC Not More
    Than 15.00
  • Accuracy for LLOQ QC level 80.00 to 120
  • Accuracy for samples other than LLOQ QC 85.00
    to 115.00
  • At least 67 of total QC samples and 50 at each
    concentration level should comply with above
    mentioned criteria of Accuracy.

20
Analysis of Study Samples
  • Analytical Run (Batch)-Samples which are
    subsequently processed without interruption in
    time by the same analyst with the same reagents
    under homogeneous conditions.
  • Acceptance Criteria
  • Reanalysis of Samples
  • Integration (Peak integration and re-integration
    described in an SOP)
  • Incurred Samples Reanalysis (ISR) - verifies the
    reliability of the reported study sample analyte
    concentrations -Two concentration levels around
    the expected Cmax in the elimination phase
  • Analytical Report

21
Reanalysis of Samples
  • Reanalysis of Samples Possible reasons defined
    in the protocol or SOP
  • Examples
  • Batch rejected (acceptance criteria for
    calibrators/QCs not met)
  • IS response in study sample significantly
    different from calibrators/QCs
  • Improper sample injection, malfunctioning
    equipment
  • Sample concentration above ULLQ
  • Quantifiable concentrations in pre-dose samples
  • Poor chromatography
  • Not acceptable in BE studies
  • Pharmacokinetic reasons (irregular profile)

22
  • Reporting
  • Records must be produced and securely stored to
    ensure proper method validation. Based on the
    validation and bioanalytical reports, a study
    should be able to be repeated as reported.
  • Regulatory Guidelines
  • With our team of dedicated scientists, we develop
    and optimize bioanalysis methods on which we then
    perform assay validation as per FDA guidelines
    using Good Laboratory Practices (GLP), regulated
    under US 21CFR part 58 and the bioanalytical
    method validation guidance for industry.

23
Conclusion
  • As you can see, bioanalytical testing is a
    complicated endeavor that provides incredibly
    valuable information about the safety and
    efficacy of drugs in a trustworthy manner.
  • Common applications of bioanalysis performed at
    Actimus BioLab include testing drug and
    metabolite exposures, bioavailability and
    bioequivalence in various studies conducted
    during preclinical (animal) and clinical (human
    volunteers) phases of drug development.
  • For expert support in developing and
    validating bioanalytical methods for your drug
    development and research, contact us at Actimus
    BioLab about our bioanalytical laboratory
    services. We seek contentment in serving our
    clients towards their noble mission of bettering
    the current standards of treatments.

24
Thank you
Write a Comment
User Comments (0)
About PowerShow.com