SlideShare uma empresa Scribd logo
1 de 74
Analytical Method Validation


              P. Raja Abhilash, M.pharm
              (Ph.D.)
              Assistant professor
              S.R. college of pharmacy.
Introduction
   Method validation is the process of documenting /
    proving that an analytical method provides analytical
    data acceptable for the intended use.
   A pharmaceutical drug product must meet all its
    specifications through out its entire shelf-life.
   The performance of product characteristics through
    out the shelf-life must be tested by analytical method
    for the product’s chemical, physiochemical,
    microbiological and biological characteristics.
   The method of analysis used must be validated. This
    is required to ensure the product’s safety and efficacy
    through out all phases of its shelf-life.
Objective

   The main objective of analytical validation
    is to ensure that a selected analytical
    procedure will give reproducible and
    reliable results that are adequate for the
    intended purpose.
   This is applicable to all the procedure either
    pharmacopoeial or non pharmacopoeial.
Steps of method development &
                   validation
   The steps of method development & validation
    depends upon the type of method being developed,
    however the following steps are common to most
    types of projects:
          Method development plan definition
          Back ground information gathering
          Laboratory method development
          Generation of test procedure
          Methods validation protocol definition
          Laboratory method validation
          Validated test method generation
          Validation report
Types of Analytical Procedures
               to be Validated
    The required validation parameters also termed
    “analytical performance characteristics”, depends
    upon the type of analytical method.
   Pharmaceutical analytical methods are
    characterized into 5 general types
      Identification tests

      Potency assays

      Limit tests for the control of impurities

      Impurity tests- quantitative

      Specific tests.
Prerequisites for analytical method validation




Man
Man                                Machine
                                   Machine                                    Methods
                                                                              Methods
      qualified
      qualified                                                         calibrated
                                                                        calibrated       characterised
                                                                                         characterised
                                                                             robust
                                                                             robust         documented
                                                                                            documented
  skilled
  skilled                                                        qualified
                                                                 qualified         suitable
                                                                                   suitable
                                                                                                            Quality
                                                                                                            Quality
Reference
Reference                                                     Vibrations
                                                              Vibrations                Time
                                                                                        Time
                                                                                                             analy
                                                                                                             analy
standards
standards                                                                                       Analysts´
                                                                              Irradi-
                                                                              Irradi-           Analysts´
                                                                                                 support
                                                                                                              meth
                                                                                                              meth
             Tempe-
             Tempe-                                                           ations
                                                                              ations             support
      Quality rature
      Quality rature                                                    Humidity
                                                                        Humidity          Supplies
                                                                                          Supplies
terial
terial                                                       Milieu Management
                                                             Milieu Management
                                                             Six “M”s
                                                              
                   
Validation parameter          ICH   USP
Accuracy                      O     O
             Repeatibility    O     O
Precision    Interm. precision O    -
             Reproducibility O      -
Specificity or Selectivity    O     O
Detection limit               O     O
Quantitation limit            O     O
Linearity                     O     O
Range                         O     O
Robustness                    O     O
Ruggedness                    -     O
Table
Specificity / selectivity


    Is this analytical     Suppose we alter
   procedure specific       test conditions
for the drug under test?       slightly?
Specificity (1)
   Specificity is the ability to assess
    unequivocally the analyte in the presence of
    components which may be expected to be
    present.
   Typically these might include impurities,
    degradants, matrix, etc.
   Lack of specificity of an individual
    analytical procedure may be compensated
    by other supporting analytical procedure(s).
Specificity (2)
   An investigation of specificity should be conducted
    during the validation of identification tests, the
    determination of impurities and the assay.
       The procedures used to demonstrate specificity will
        depend on the intended objective of the analytical
        procedure.
   It is not always possible to demonstrate that an
    analytical procedure is specific for a particular
    analyte (complete discrimination).
       In this case a combination of two or more analytical
        procedures is recommended to achieve the necessary
        level of discrimination.
Identification
   Suitable identification tests should be able to
    discriminate between compounds of closely related
    structures which are likely to be present.
       The discrimination of a procedure may be confirmed by
        obtaining positive results (perhaps by comparison with
        a known reference material) from samples containing
        the analyte, coupled with negative results from samples
        which do not contain the analyte.
       In addition, the identification test may be applied to
        materials structurally similar to or closely related to the
        analyte to confirm that a positive response is not
        obtained.
Assay and Impurity Test(s)
   For chromatographic procedures, representative
    chromatograms should be used to demonstrate specificity
    and individual components should be appropriately
    labelled.
   Critical separations in chromatography should be
    investigated at an appropriate level.
       For critical separations, specificity can be demonstrated by the
        resolution of the two components which elute closest to each other.
   In cases where a non-specific assay is used, other
    supporting analytical procedures should be used to
    demonstrate overall specificity.
       For example, where a titration is adopted to assay the drug
        substance for release, the combination of the assay and a suitable
        test for impurities can be used.
Impurities are available
   For the assay , this should involve demonstration
    of the discrimination of the analyte in the presence
    of impurities and/or excipients;
   practically, this can be done by spiking pure
    substances (drug substance or drug product) with
    appropriate levels of impurities and/or excipients
    and demonstrating that the assay result is
    unaffected by the presence of these materials
Impurities are not available
   specificity may be demonstrated by comparing the
    test results of samples containing impurities or
    degradation products to a second well-
    characterized procedure e.g.: spharmacopoeial
    method or other validated analytical procedure
    (independent procedure).
   As appropriate, this should include samples stored
    under relevant stress conditions:
       light, heat, humidity, acid/base hydrolysis and
        oxidation.
Linearity and Range
‘Know that it’s a straight line’
              vs
‘For what concentrations is it a
  straight line’
Linearity and Range

Concentration
  mg/mL




                              Response
      ‘Know that it’s a straight line’ versus ‘For what
       concentrations is it a straight line’
            Is it a straight line between 0.4 & 0.6 mg/mL?
            Over what range is it a straight line?
            Answer: approx 0.25-0.70 mg/mL
LINEARITY (1)
   A linear relationship should be evaluated
    across the range of the analytical
    procedure.
   It may be demonstrated directly on the
    drug substance by:
       dilution of a standard stock solution and/or
       separate weighing of synthetic mixtures of the
        drug product components using the proposed
        procedure.
LINEARITY (2)
   Linearity should be evaluated by visual inspection of a plot of
    signals as a function of analyte concentration or content.
   If there is a linear relationship, test results should be
    evaluated by appropriate statistical methods, for example, by
    calculation of a regression line by the method of least squares.
   In some cases, to obtain linearity between assays and sample
    concentrations, the test data may need to be subjected to a
    mathematical transformation prior to the regression analysis.
    Data from the regression line itself may be helpful to provide
    mathematical estimates of the degree of linearity.
   For the establishment of linearity, a minimum of 5
    concentrations is recommended.
Example
   Seven solutions containing different
    concentrations (0.280 – 0.520) mg/ml of
    ketotifen fumarate in tablets were assayed
    using HPLC
   The results were evaluated statistically and
    the results shown on the following slide
Example (continued)
Concentration of ketotifen fumarate   Area detected   Acceptance
       mg/ml                   %                      criteria


        0.280                 70         1473566
        0.320                 80         1677013
        0.360                 90         1904848
        0.400                 100        2091215
        0.440                 110        2293647
        0.480                 120        2518976
        0.520                 130        2670144

Regression: y = ax + b                                0.998 – 1.002
            a = 5055766.964
            b = 67608.786
            r2 = 0.9984
RANGE
   The specified range is normally derived from
    linearity studies and depends on the intended
    application of the procedure.
   It is established by confirming that the analytical
    procedure provides an acceptable degree of
    linearity, accuracy and precision when applied to
    samples containing amounts of analyte within or
    at the extremes of the specified range of the
    analytical procedure.
Minimum Specified Ranges
   for the assay of a drug substance or a finished
    (drug) product: normally from 80 - 120 % of the test
    concentration
   for content uniformity, covering a minimum of
    70 - 130 % of the test concentration
   for dissolution testing: +/-20 % over the
    specified range;
    e.g., if the specifications for a controlled released
    product cover a region from 20%, after 1 hour, up to
    90%, after 24 hours, the validated range would be 0-
    110% of the label claim
Accuracy vs precision
            What you
                would
            like
             to see!
Accuracy vs precision


                Poor accuracy
                Good precision
Accuracy vs precision


              Poor precision
              Good accuracy
Accuracy vs precision
What would you
 call this?



                    Totally hopeless!
                    Poor precision
                    Poor accuracy
So what definitions do these
concepts lead us to in the
context of assay validation?
ACCURACY (1)
   The accuracy of an analytical procedure
    expresses the closeness of agreement
    between the value which is accepted either
    as a conventional true value or an accepted
    reference value and the value found. This is
    sometimes termed trueness.
ACCURACY (2)
Assay of Drug Product:
a) Application of the analytical procedure to synthetic
   mixtures of the drug product components to which known
   quantities of the drug substance to be analysed have been
   added (standard addition method);
b) In cases where it is impossible to obtain samples of all drug
   product components, it may be acceptable either to:
      add known quantities of the analyte to the drug product or
      to compare the results obtained from a second, well characterized
       procedure, the accuracy of which is stated and/or defined
       (independent procedure)
c) Accuracy may be inferred once precision, linearity and
   specificity have been established.
ACCURACY (3)
Impurities (Quantitation):
 Accuracy should be assessed on samples (drug

  substance/drug product) spiked with known amounts of
  impurities.
 In cases where it is impossible to obtain samples of certain

  impurities and/or degradation products, it is considered
  acceptable to compare results obtained by an independent
  procedure. The response factor of the drug substance can
  be used.
 It should be clear how the individual or total impurities

  are to be determined e.g., weight/weight or area percent, in
  all cases with respect to the major analyte.
Recommended Data
   Accuracy should be assessed using a min. of
    9 determinations over a min. of 3
    concentration levels covering the specified
    range (e.g. 3 concentrations/3 replicates each of
    the total analytical procedure).
   Accuracy should be reported as:
       % recovery by the assay of known added amount of
        analyte in the sample or as
       the difference between the mean and the
        accepted true value together with the
        confidence intervals
Example:

   Nine solutions containing different
    concentrations of ketotifen fumarate
    reference standard added to ketotifen tablet
    were assayed
Example (continued):
Conc. of ketotifen fumarate     Area            Recovery   Acceptance
   mg/ml            %          detected           (%)       Criteria


   0.280            70         1473566            99.32
   0.320            80         1677013            99.48
   0.360            90         1904848           100.94
   0.380            95         1905862           100.51
   0.400           100         2091215           100.06
   0.420           105         2180374           100.03
   0.440           110         2293647           100.07
   0.480           120         2518976           101.01
   0.520           130         2670144            98.99

Mean (recovery)                     : 100.04               98.0–102.0 %
Standard deviation                  : 0.699
Relative standard deviation (RSD)   : 0.699 %                 <2%
PRECISION
   The precision of an analytical procedure expresses the
    closeness of agreement (degree of scatter) between a series
    of measurements obtained from multiple sampling of the
    same homogeneous sample under the prescribed
    conditions.
   Precision may be considered at three levels:
       repeatability,
       intermediate precision and
       reproducibility.
   Precision should be investigated using homogeneous,
    authentic samples. However, if it is not possible to obtain a
    homogeneous sample it may be investigated using
    artificially prepared samples or a sample solution.
   The precision of an analytical procedure is usually
    expressed as the variance, standard deviation or
    coefficient of variation of a series of measurements.
Repeatability (1)

   Repeatability expresses the precision under
    the same operating conditions over a short
    interval of time.
   Repeatability is also termed intra-assay
    precision.
Repeatability (2)
   Repeatability should be assessed using:
    a) a minimum of 9 determinations
    covering the specified range for the
    procedure (e.g. 3 concentrations/3
    replicates each) or
    b) a minimum of 6 determinations at
    100% of the test concentration.
Intermediate precision
   Intermediate precision expresses within-laboratories
    variations: different days, different analysts,
    different equipment, etc.
   The extent to which intermediate precision should be
    established depends on the circumstances under which the
    procedure is intended to be used.
   The applicant should establish the effects of random events
    on the precision of the analytical procedure.
   Typical variations to be studied include days, analysts,
    equipment, etc. It is not considered necessary to study
    these effects individually. The use of an experimental
    design (matrix) is encouraged.
Reproducibility
   Reproducibility is assessed by means of an
    inter-laboratory trial.
   Reproducibility should be considered in
    case of the standardization of an analytical
    procedure, for instance, for inclusion of
    procedures in pharmacopoeias.
   These data are not part of the marketing
    authorization dossier.
Recommended Data

   The standard deviation, relative
    standard deviation (coefficient of
    variation) and confidence interval should be
    reported for each type of precision
    investigated.
Example
   The active ingredient, ketotifen fumarate,
    in tablets was assayed seven times using
    HPLC and the reference standard
Example (continued)
 Sample no.        Concentration (mg/ml)        Area detected
      1                     0.4                    1902803
      2                     0.4                    1928083
      3                     0.4                    1911457
      4                     0.4                    1915897
      5                     0.4                    1913312
      6                     0.4                    1897702
      7                     0.4                    1907019

Mean                                       :   1910896
Standard deviation                         :   9841.78
Relative standard deviation (RSD)          :   0.515 %

Acceptance criteria:
Relative standard deviation (RSD): not more than 2 %
Detection limit vs
  Quantitation limit

 ‘Know that it’s there’
          vs
‘Know how much is there’
Detection limit
       (means)
Is any of it present?




        Is it there?
Quantitation limit
How much of it is present???




  How much of it is there?
DETECTION LIMIT
   The detection limit of an individual
    analytical procedure is the lowest amount of
    analyte in a sample which can be detected
    but not necessarily quantitated as an exact
    value
   Several approaches for determining the
    detection limit are possible, depending on
    whether the procedure is a non-
    instrumental or instrumental.
Based on Visual Evaluation
   Visual evaluation may be used for non-
    instrumental methods but may also be used
    with instrumental methods.
   The detection limit is determined by the
    analysis of samples with known
    concentrations of analyte and by
    establishing the minimum level at which the
    analyte can be reliably detected .
Based on Signal-to-Noise
   This approach can only be applied to analytical
    procedures which exhibit baseline noise.
   Determination of the signal-to-noise ratio is
    performed by comparing measured signals from
    samples with known low concentrations of analyte
    with those of blank samples and establishing the
    minimum concentration at which the analyte can
    be reliably detected.
   A signal-to-noise ratio between 3:1 or 2:1 is
    generally considered acceptable for estimating the
    detection limit.
Based on the Standard Deviation of
   the Response and the Slope
The detection limit (DL) may be expressed
as:
DL = 3.3 σ /S
where σ = the standard deviation of the
response
S = the slope of the calibration curve
The slope S may be estimated from the
calibration curve of the analyte.
Estimate of σ
   Based on the Standard Deviation of the Blank
       Measurement of the magnitude of analytical
        background response is performed by analyzing an
        appropriate number of blank samples and calculating
        the standard deviation of these responses
   Based on the Calibration Curve
       A specific calibration curve should be studied using
        samples containing an analyte in the range of DL.
       The residual standard deviation of a regression line or
        the standard deviation of y-intercepts of regression lines
        may be used as the standard deviation.
Recommended Data
   The detection limit and the method used for
    determining the detection limit should be
    presented.
   If DL is determined based on visual evaluation or
    based on signal to noise ratio, the presentation of
    the relevant chromatograms is considered
    acceptable for justification.
   In cases where an estimated value for the detection
    limit is obtained by calculation or extrapolation,
    this estimate may subsequently be validated by the
    independent analysis of a suitable number of
    samples known to be near or prepared at the
    detection limit
QUANTITATION LIMIT
   The quantitation limit of an individual analytical
    procedure is the lowest amount of analyte in a
    sample which can be quantitatively determined
    with suitable precision and accuracy.
   The quantitation limit is a parameter of
    quantitative assays for low levels of compounds in
    sample matrices, and is used particularly for the
    determination of impurities and/or degradation
    products.
   Several approaches for determining the
    quantitation limit are possible, depending on
    whether the procedure is a non-instrumental or
    instrumental.
Based on Visual Evaluation
   Visual evaluation may be used for non-
    instrumental methods but may also be used
    with instrumental methods.
   The quantitation limit is generally
    determined by the analysis of samples with
    known concentrations of analyte and by
    establishing the minimum level at which the
    analyte can be quantified with acceptable
    accuracy and precision.
Based on
         Signal-to-Noise Approach
   This approach can only be applied to analytical
    procedures that exhibit baseline noise.
   Determination of the signal-to-noise ratio is
    performed by comparing measured signals from
    samples with known low concentrations of analyte
    with those of blank samples and by establishing
    the minimum concentration at which the analyte
    can be reliably quantified.
   A typical signal-to-noise ratio is 10:1.
Based on the Standard Deviation of
   the Response and the Slope
   The quantitation limit (QL) may be
    expressed as:
    QL = 10 σ /S
    where σ = the standard deviation of the
    response
    S = the slope of the calibration curve
   The slope S may be estimated from the
    calibration curve of the analyte.
Estimate of σ
   Based on Standard Deviation of the Blank
       Measurement of the magnitude of analytical
        background response is performed by analyzing an
        appropriate number of blank samples and calculating
        the standard deviation of these responses.
   Based on the Calibration Curve
       A specific calibration curve should be studied using
        samples, containing an analyte in the range of QL.
       The residual standard deviation of a regression line or
        the standard deviation of y-intercepts of regression lines
        may be used as the standard deviation.
Recommended Data
   The quantitation limit and the method used
    for determining the quantitation limit
    should be presented.
   The limit should be subsequently validated
    by the analysis of a suitable number of
    samples known to be near or prepared at
    the quantitation limit.
Robustness
Small changes do not affect
 the parameters of the
 assay
ROBUSTNESS
   The robustness of an analytical procedure is a measure of
    its capacity to remain unaffected by small, but deliberate
    variations in method parameters and provides an
    indication of its reliability during normal usage.
   The evaluation of robustness should be considered during
    the development phase and depends on the type of
    procedure under study.
   If measurements are susceptible to variations in analytical
    conditions, the analytical conditions should be suitably
    controlled or a precautionary statement should be included
    in the procedure.
   One consequence of the evaluation of robustness should be
    that a series of system suitability parameters (e.g.,
    resolution test) is established to ensure that the validity of
    the analytical procedure is maintained whenever used.
Typical Variations
 stability of analytical solutions,
 extraction time

Liquid chromatography:
 influence of variations of pH in a mobile phase,

 influence of variations in mobile phase composition,

 different columns (different lots and/or suppliers),

 temperature,

 flow rate.

Gas chromatography:
 different columns (different lots and/or suppliers),

 temperature,

 flow rate.
SYSTEM SUITABILITY TESTING
   System suitability testing is an integral part of
    many analytical procedures.
   The tests are based on the concept that the
    equipment, electronics, analytical operations and
    samples to be analyzed constitute an integral
    system that can be evaluated as such.
   System suitability test parameters to be
    established for a particular procedure depend on
    the type of procedure being validated. They are
    especially important in the case of
    chromatographic methods.
System Suitability in
                Chromatography
   To verify that the resolution and reproducibility of the
    chromatographic system are adequate for the analysis to
    be done
   The resolution, R, is specified to ensure that closely eluting
    compounds are resolved from each other
   Replicate injections of a standard preparation are
    compared to ascertain whether requirements for precision
    are met
   The tailing factor, T, has to meet a certain requirement,
    because as peak asymmetry increases, integration, and
    hence precision, becomes less reliable
Evaluating validation data for an
             HPLC procedure

   Here are some suggestions………
   But please note!
-   The slides that follow do not represent requirements; they
    are suggestions.
-   There is more than one way to do this!
-   Use judgement.

   If you are unsure, consult with experienced analysts!!
Specificity
                     (selectivity)

Use some or all of these procedures:
-   Add a synthetic mixture of excipients to the sample &
    check whether the assay result for the drug is the same
-   Add some known impurities to the test sample & check
    whether they are resolved (separated from) the drug
-   Forcably degrade the active & test whether degradants are
    separated from the intact drug
-   Assess peak purity by diode array
Linearity
-   Minimum of 5 concentrations
-   r2 >0.99 if possible
-   Intercept NMT ±2% of response of 100% the
    working concentration
-   Confirm accuracy & precision over the
    required range
Accuracy
-   Generally within +2%
    -   Recoveries after spiking, or
    -   Comparison with ‘well-established’ methods & by
        inference
-   Arguably can be up to +10% for
    related substances
-   What is known about the reference
    standard?
Precision
               - repeatability
   System repeatability   %CV (of detector
                           response) <2.0% for 6
                           injections

   Method repeatability   %CV <2.0%
                           and accuracy
                           should be within 2%
Precision -     intermediate
          [= ruggedness USP]
-   Use same complete analytical procedure for
    comparisons
-   Compare results across different analysts, days,
    equipment
-   Means preferably within 2%
-   Compare %CV with that for method repeatability
Precision
          - reproducibility
-   This is not normally a component of a dossier for
    an application to register, but if you do have to
    evaluate these data then……
-   For interlab comparisons
     - Means should preferably be within 2%

     - Compare the %CV with that for method

       repeatability
     - Can use an F test, normally with 95%

       confidence
Limit of detection
-   Use some or all of these procedures:
-   - Visual evaluation: A clear & symmetrical peak is
    visible
-   Signal to noise ratio of 3:1 or 2:1
-   Based on statistical information:
    -   Detection limit =
    -   3.3 x (std dev at that concentration)
    -              slope
Limit of quantitation
-   Use some or all of these procedures:
-   ‘Visual’ evaluation: A clear & symmetrical peak is
    visible
-   Signal to noise ratio of 10:1
-   Based on statistical information:
    -   Detection limit =
    -   10 x (std dev at that concentration)
    -              slope
Robustness
-   Use some or all of these procedures:
-   Compare results after altering HPLC parameters,
    eg mobile phase composition, buffer composition,
    pH, column type, flow rate:
    -   NMT ± 2% difference in assay
-   Compare results after storage of test solution, eg
    for 24h at say 250C
    -   NMT ± 2% difference in assay
Evaluation of analytical validation data
  The objective of the analytical procedure

  The analytical technique

                    Item                       Data provided by applicant   Acceptable or not? (add comments if
                                                      (very briefly)         necessary, & reasons if unacceptable)
  Is a chromatogram, spectrum or
        similar provided?
  S




  Linearity

  Range

  Accuracy

  Precision: Repeatability

  Precision: Intermediate

  Precision: Reproducibility

  Detection limit

  Quantitation limit

  Robustness

 System suitability (if necessary)
     Are the data concerning analytical validation satisfactory? YES/NO
 DataIfon therecommended questions to the applicant appear in
        NO, reference standard
……………………………………………………………………
 Other evaluator comments:
     (eg page number below, or draft letter to the company on page……)
Thank you

Mais conteúdo relacionado

Mais procurados

ICH Q2 Analytical Method Validation
ICH Q2  Analytical Method ValidationICH Q2  Analytical Method Validation
ICH Q2 Analytical Method ValidationNaila Kanwal
 
Stability testing protocols
Stability testing protocolsStability testing protocols
Stability testing protocolsMehulJain143
 
Qualification & Validation
Qualification & ValidationQualification & Validation
Qualification & ValidationICHAPPS
 
PHARMACEUTICAL VALIDATION
 PHARMACEUTICAL  VALIDATION PHARMACEUTICAL  VALIDATION
PHARMACEUTICAL VALIDATIONSACHIN C P
 
ICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESHEALY LAD
 
Impurities in Drug Substance & in Drug Product
Impurities in Drug Substance & in Drug ProductImpurities in Drug Substance & in Drug Product
Impurities in Drug Substance & in Drug ProductKamal Ambalia
 
Manufacturing operation and controls
Manufacturing operation and controls Manufacturing operation and controls
Manufacturing operation and controls sachin pawar
 
Impurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek JainImpurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek JainVivek Jain
 
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLC
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLCQUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLC
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLCAnupriyaNR
 
Ipqc tests for tablet
Ipqc tests for tabletIpqc tests for tablet
Ipqc tests for tabletMalay Jivani
 
Ich guidelines for stability studies 1
Ich guidelines for stability studies 1Ich guidelines for stability studies 1
Ich guidelines for stability studies 1priyanka odela
 
STABILITY TESTING DURING PRODUCT DEVELOPMENT
STABILITY TESTING DURING PRODUCT DEVELOPMENTSTABILITY TESTING DURING PRODUCT DEVELOPMENT
STABILITY TESTING DURING PRODUCT DEVELOPMENTAmruta Balekundri
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validationSHIVANEE VYAS
 
validation of analytical method used in cleaning
validation of analytical method used in cleaningvalidation of analytical method used in cleaning
validation of analytical method used in cleaningKUNDLAJAYALAKSHMI
 
Ipqc for parenterals
Ipqc for parenteralsIpqc for parenterals
Ipqc for parenteralsceutics1315
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instrumentsFaris ameen
 
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage Forms
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage FormsIn Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage Forms
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage FormsSagar Savale
 

Mais procurados (20)

Validation parameters
Validation parametersValidation parameters
Validation parameters
 
ICH Q2 Analytical Method Validation
ICH Q2  Analytical Method ValidationICH Q2  Analytical Method Validation
ICH Q2 Analytical Method Validation
 
Stability testing protocols
Stability testing protocolsStability testing protocols
Stability testing protocols
 
Qualification & Validation
Qualification & ValidationQualification & Validation
Qualification & Validation
 
PHARMACEUTICAL VALIDATION
 PHARMACEUTICAL  VALIDATION PHARMACEUTICAL  VALIDATION
PHARMACEUTICAL VALIDATION
 
Stability studies
Stability studies Stability studies
Stability studies
 
ICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIESICH GUIDELINE SPECIFIC WITH Q SERIES
ICH GUIDELINE SPECIFIC WITH Q SERIES
 
Impurities in Drug Substance & in Drug Product
Impurities in Drug Substance & in Drug ProductImpurities in Drug Substance & in Drug Product
Impurities in Drug Substance & in Drug Product
 
Manufacturing operation and controls
Manufacturing operation and controls Manufacturing operation and controls
Manufacturing operation and controls
 
Impurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek JainImpurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek Jain
 
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLC
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLCQUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLC
QUALIFICATION OF UV-VISIBLE SPECTROPHOTOMETER, FTIR, DSC, HPLC
 
Ipqc tests for tablet
Ipqc tests for tabletIpqc tests for tablet
Ipqc tests for tablet
 
Ich guidelines for stability studies 1
Ich guidelines for stability studies 1Ich guidelines for stability studies 1
Ich guidelines for stability studies 1
 
Residual solvents
Residual solventsResidual solvents
Residual solvents
 
STABILITY TESTING DURING PRODUCT DEVELOPMENT
STABILITY TESTING DURING PRODUCT DEVELOPMENTSTABILITY TESTING DURING PRODUCT DEVELOPMENT
STABILITY TESTING DURING PRODUCT DEVELOPMENT
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
validation of analytical method used in cleaning
validation of analytical method used in cleaningvalidation of analytical method used in cleaning
validation of analytical method used in cleaning
 
Ipqc for parenterals
Ipqc for parenteralsIpqc for parenterals
Ipqc for parenterals
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
 
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage Forms
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage FormsIn Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage Forms
In Process Quality Control Tests (IPQC) For Parenteral or Sterile Dosage Forms
 

Destaque

Analytical mehod validation explained sadasiva
Analytical mehod validation explained sadasivaAnalytical mehod validation explained sadasiva
Analytical mehod validation explained sadasivaSada Siva Rao Maddiguntla
 
Related Substances-Method Validation-PPT_slide
Related Substances-Method Validation-PPT_slideRelated Substances-Method Validation-PPT_slide
Related Substances-Method Validation-PPT_slideBhanu Prakash N
 
Method Development and Method Validation for the estimation of Valganciclovir...
Method Development and Method Validation for the estimation of Valganciclovir...Method Development and Method Validation for the estimation of Valganciclovir...
Method Development and Method Validation for the estimation of Valganciclovir...google
 
Analytical method development,validation by uv spectroscopy
Analytical method development,validation by uv spectroscopyAnalytical method development,validation by uv spectroscopy
Analytical method development,validation by uv spectroscopythota lakshmi bhavani
 
Assay-Method validation-PPT _slide
Assay-Method validation-PPT _slideAssay-Method validation-PPT _slide
Assay-Method validation-PPT _slideBhanu Prakash N
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validationShivalingam Anugu
 
Analytical method validation workshop
Analytical method validation workshopAnalytical method validation workshop
Analytical method validation workshopSiham Abdallaha
 
Analytical Method Validation as per ICH vs USP
Analytical Method Validation as per ICH vs USPAnalytical Method Validation as per ICH vs USP
Analytical Method Validation as per ICH vs USPKushal Shah
 
Calibration and validation of analytical instruments
Calibration and validation of analytical instrumentsCalibration and validation of analytical instruments
Calibration and validation of analytical instrumentsSolairajan A
 
Analytical method validation by manoj ingale(best ppts)
Analytical method validation by manoj ingale(best ppts)Analytical method validation by manoj ingale(best ppts)
Analytical method validation by manoj ingale(best ppts)Indus Biotech Pvt.Ltd.
 
analytical method validation and validation of hplc
analytical method validation and validation of hplcanalytical method validation and validation of hplc
analytical method validation and validation of hplcvenkatesh thota
 
Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method ValidationStefan Holt
 
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR Dr. Ravi Sankar
 

Destaque (16)

Analytical mehod validation explained sadasiva
Analytical mehod validation explained sadasivaAnalytical mehod validation explained sadasiva
Analytical mehod validation explained sadasiva
 
Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
 
Related Substances-Method Validation-PPT_slide
Related Substances-Method Validation-PPT_slideRelated Substances-Method Validation-PPT_slide
Related Substances-Method Validation-PPT_slide
 
Method Development and Method Validation for the estimation of Valganciclovir...
Method Development and Method Validation for the estimation of Valganciclovir...Method Development and Method Validation for the estimation of Valganciclovir...
Method Development and Method Validation for the estimation of Valganciclovir...
 
Analytical method development,validation by uv spectroscopy
Analytical method development,validation by uv spectroscopyAnalytical method development,validation by uv spectroscopy
Analytical method development,validation by uv spectroscopy
 
Assay-Method validation-PPT _slide
Assay-Method validation-PPT _slideAssay-Method validation-PPT _slide
Assay-Method validation-PPT _slide
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
Analytical method validation
Analytical method validation Analytical method validation
Analytical method validation
 
Analytical method validation workshop
Analytical method validation workshopAnalytical method validation workshop
Analytical method validation workshop
 
validation
validationvalidation
validation
 
Analytical Method Validation as per ICH vs USP
Analytical Method Validation as per ICH vs USPAnalytical Method Validation as per ICH vs USP
Analytical Method Validation as per ICH vs USP
 
Calibration and validation of analytical instruments
Calibration and validation of analytical instrumentsCalibration and validation of analytical instruments
Calibration and validation of analytical instruments
 
Analytical method validation by manoj ingale(best ppts)
Analytical method validation by manoj ingale(best ppts)Analytical method validation by manoj ingale(best ppts)
Analytical method validation by manoj ingale(best ppts)
 
analytical method validation and validation of hplc
analytical method validation and validation of hplcanalytical method validation and validation of hplc
analytical method validation and validation of hplc
 
Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
 
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR
ANALYTICAL METHOD VALIDATION BY P.RAVISANKAR
 

Último

Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdf
Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdfLippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdf
Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdfSreeja Cherukuru
 
Primary headache and facial pain. (2024)
Primary headache and facial pain. (2024)Primary headache and facial pain. (2024)
Primary headache and facial pain. (2024)Mohamed Rizk Khodair
 
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...MehranMouzam
 
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptx
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptxPERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptx
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptxdrashraf369
 
History and Development of Pharmacovigilence.pdf
History and Development of Pharmacovigilence.pdfHistory and Development of Pharmacovigilence.pdf
History and Development of Pharmacovigilence.pdfSasikiranMarri
 
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdf
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdfMedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdf
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdfSasikiranMarri
 
PULMONARY EDEMA AND ITS MANAGEMENT.pdf
PULMONARY EDEMA AND  ITS  MANAGEMENT.pdfPULMONARY EDEMA AND  ITS  MANAGEMENT.pdf
PULMONARY EDEMA AND ITS MANAGEMENT.pdfDolisha Warbi
 
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...Wessex Health Partners
 
Informed Consent Empowering Healthcare Decision-Making.pptx
Informed Consent Empowering Healthcare Decision-Making.pptxInformed Consent Empowering Healthcare Decision-Making.pptx
Informed Consent Empowering Healthcare Decision-Making.pptxSasikiranMarri
 
Presentation on General Anesthetics pdf.
Presentation on General Anesthetics pdf.Presentation on General Anesthetics pdf.
Presentation on General Anesthetics pdf.Prerana Jadhav
 
Giftedness: Understanding Everyday Neurobiology for Self-Knowledge
Giftedness: Understanding Everyday Neurobiology for Self-KnowledgeGiftedness: Understanding Everyday Neurobiology for Self-Knowledge
Giftedness: Understanding Everyday Neurobiology for Self-Knowledgeassessoriafabianodea
 
maternal mortality and its causes and how to reduce maternal mortality
maternal mortality and its causes and how to reduce maternal mortalitymaternal mortality and its causes and how to reduce maternal mortality
maternal mortality and its causes and how to reduce maternal mortalityhardikdabas3
 
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdf
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdfSGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdf
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdfHongBiThi1
 
Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.ANJALI
 
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic Analysis
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic AnalysisVarSeq 2.6.0: Advancing Pharmacogenomics and Genomic Analysis
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic AnalysisGolden Helix
 
world health day presentation ppt download
world health day presentation ppt downloadworld health day presentation ppt download
world health day presentation ppt downloadAnkitKumar311566
 
systemic bacteriology (7)............pptx
systemic bacteriology (7)............pptxsystemic bacteriology (7)............pptx
systemic bacteriology (7)............pptxEyobAlemu11
 
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara Rajendran
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara RajendranMusic Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara Rajendran
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara RajendranTara Rajendran
 
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...Badalona Serveis Assistencials
 

Último (20)

Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdf
Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdfLippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdf
Lippincott Microcards_ Microbiology Flash Cards-LWW (2015).pdf
 
Primary headache and facial pain. (2024)
Primary headache and facial pain. (2024)Primary headache and facial pain. (2024)
Primary headache and facial pain. (2024)
 
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...
Study on the Impact of FOCUS-PDCA Management Model on the Disinfection Qualit...
 
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptx
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptxPERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptx
PERFECT BUT PAINFUL TKR -ROLE OF SYNOVECTOMY.pptx
 
History and Development of Pharmacovigilence.pdf
History and Development of Pharmacovigilence.pdfHistory and Development of Pharmacovigilence.pdf
History and Development of Pharmacovigilence.pdf
 
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdf
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdfMedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdf
MedDRA-A-Comprehensive-Guide-to-Standardized-Medical-Terminology.pdf
 
PULMONARY EDEMA AND ITS MANAGEMENT.pdf
PULMONARY EDEMA AND  ITS  MANAGEMENT.pdfPULMONARY EDEMA AND  ITS  MANAGEMENT.pdf
PULMONARY EDEMA AND ITS MANAGEMENT.pdf
 
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...
Wessex Health Partners Wessex Integrated Care, Population Health, Research & ...
 
Informed Consent Empowering Healthcare Decision-Making.pptx
Informed Consent Empowering Healthcare Decision-Making.pptxInformed Consent Empowering Healthcare Decision-Making.pptx
Informed Consent Empowering Healthcare Decision-Making.pptx
 
Presentation on General Anesthetics pdf.
Presentation on General Anesthetics pdf.Presentation on General Anesthetics pdf.
Presentation on General Anesthetics pdf.
 
Giftedness: Understanding Everyday Neurobiology for Self-Knowledge
Giftedness: Understanding Everyday Neurobiology for Self-KnowledgeGiftedness: Understanding Everyday Neurobiology for Self-Knowledge
Giftedness: Understanding Everyday Neurobiology for Self-Knowledge
 
maternal mortality and its causes and how to reduce maternal mortality
maternal mortality and its causes and how to reduce maternal mortalitymaternal mortality and its causes and how to reduce maternal mortality
maternal mortality and its causes and how to reduce maternal mortality
 
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdf
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdfSGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdf
SGK HÓA SINH NĂNG LƯỢNG SINH HỌC 2006.pdf
 
Epilepsy
EpilepsyEpilepsy
Epilepsy
 
Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.Statistical modeling in pharmaceutical research and development.
Statistical modeling in pharmaceutical research and development.
 
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic Analysis
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic AnalysisVarSeq 2.6.0: Advancing Pharmacogenomics and Genomic Analysis
VarSeq 2.6.0: Advancing Pharmacogenomics and Genomic Analysis
 
world health day presentation ppt download
world health day presentation ppt downloadworld health day presentation ppt download
world health day presentation ppt download
 
systemic bacteriology (7)............pptx
systemic bacteriology (7)............pptxsystemic bacteriology (7)............pptx
systemic bacteriology (7)............pptx
 
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara Rajendran
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara RajendranMusic Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara Rajendran
Music Therapy's Impact in Palliative Care| IAPCON2024| Dr. Tara Rajendran
 
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...
Presentació "Real-Life VR Integration for Mild Cognitive Impairment Rehabilit...
 

analytical method validation

  • 1. Analytical Method Validation P. Raja Abhilash, M.pharm (Ph.D.) Assistant professor S.R. college of pharmacy.
  • 2. Introduction  Method validation is the process of documenting / proving that an analytical method provides analytical data acceptable for the intended use.  A pharmaceutical drug product must meet all its specifications through out its entire shelf-life.  The performance of product characteristics through out the shelf-life must be tested by analytical method for the product’s chemical, physiochemical, microbiological and biological characteristics.  The method of analysis used must be validated. This is required to ensure the product’s safety and efficacy through out all phases of its shelf-life.
  • 3. Objective  The main objective of analytical validation is to ensure that a selected analytical procedure will give reproducible and reliable results that are adequate for the intended purpose.  This is applicable to all the procedure either pharmacopoeial or non pharmacopoeial.
  • 4. Steps of method development & validation  The steps of method development & validation depends upon the type of method being developed, however the following steps are common to most types of projects:  Method development plan definition  Back ground information gathering  Laboratory method development  Generation of test procedure  Methods validation protocol definition  Laboratory method validation  Validated test method generation  Validation report
  • 5. Types of Analytical Procedures to be Validated  The required validation parameters also termed “analytical performance characteristics”, depends upon the type of analytical method.  Pharmaceutical analytical methods are characterized into 5 general types  Identification tests  Potency assays  Limit tests for the control of impurities  Impurity tests- quantitative  Specific tests.
  • 6. Prerequisites for analytical method validation Man Man Machine Machine Methods Methods qualified qualified calibrated calibrated characterised characterised robust robust documented documented skilled skilled qualified qualified suitable suitable Quality Quality Reference Reference Vibrations Vibrations Time Time analy analy standards standards Analysts´ Irradi- Irradi- Analysts´ support meth meth Tempe- Tempe- ations ations support Quality rature Quality rature Humidity Humidity Supplies Supplies terial terial Milieu Management Milieu Management Six “M”s  
  • 7. Validation parameter ICH USP Accuracy O O Repeatibility O O Precision Interm. precision O - Reproducibility O - Specificity or Selectivity O O Detection limit O O Quantitation limit O O Linearity O O Range O O Robustness O O Ruggedness - O
  • 9. Specificity / selectivity Is this analytical Suppose we alter procedure specific test conditions for the drug under test? slightly?
  • 10. Specificity (1)  Specificity is the ability to assess unequivocally the analyte in the presence of components which may be expected to be present.  Typically these might include impurities, degradants, matrix, etc.  Lack of specificity of an individual analytical procedure may be compensated by other supporting analytical procedure(s).
  • 11. Specificity (2)  An investigation of specificity should be conducted during the validation of identification tests, the determination of impurities and the assay.  The procedures used to demonstrate specificity will depend on the intended objective of the analytical procedure.  It is not always possible to demonstrate that an analytical procedure is specific for a particular analyte (complete discrimination).  In this case a combination of two or more analytical procedures is recommended to achieve the necessary level of discrimination.
  • 12. Identification  Suitable identification tests should be able to discriminate between compounds of closely related structures which are likely to be present.  The discrimination of a procedure may be confirmed by obtaining positive results (perhaps by comparison with a known reference material) from samples containing the analyte, coupled with negative results from samples which do not contain the analyte.  In addition, the identification test may be applied to materials structurally similar to or closely related to the analyte to confirm that a positive response is not obtained.
  • 13. Assay and Impurity Test(s)  For chromatographic procedures, representative chromatograms should be used to demonstrate specificity and individual components should be appropriately labelled.  Critical separations in chromatography should be investigated at an appropriate level.  For critical separations, specificity can be demonstrated by the resolution of the two components which elute closest to each other.  In cases where a non-specific assay is used, other supporting analytical procedures should be used to demonstrate overall specificity.  For example, where a titration is adopted to assay the drug substance for release, the combination of the assay and a suitable test for impurities can be used.
  • 14. Impurities are available  For the assay , this should involve demonstration of the discrimination of the analyte in the presence of impurities and/or excipients;  practically, this can be done by spiking pure substances (drug substance or drug product) with appropriate levels of impurities and/or excipients and demonstrating that the assay result is unaffected by the presence of these materials
  • 15. Impurities are not available  specificity may be demonstrated by comparing the test results of samples containing impurities or degradation products to a second well- characterized procedure e.g.: spharmacopoeial method or other validated analytical procedure (independent procedure).  As appropriate, this should include samples stored under relevant stress conditions:  light, heat, humidity, acid/base hydrolysis and oxidation.
  • 16. Linearity and Range ‘Know that it’s a straight line’ vs ‘For what concentrations is it a straight line’
  • 17. Linearity and Range Concentration mg/mL Response  ‘Know that it’s a straight line’ versus ‘For what concentrations is it a straight line’  Is it a straight line between 0.4 & 0.6 mg/mL?  Over what range is it a straight line?  Answer: approx 0.25-0.70 mg/mL
  • 18. LINEARITY (1)  A linear relationship should be evaluated across the range of the analytical procedure.  It may be demonstrated directly on the drug substance by:  dilution of a standard stock solution and/or  separate weighing of synthetic mixtures of the drug product components using the proposed procedure.
  • 19. LINEARITY (2)  Linearity should be evaluated by visual inspection of a plot of signals as a function of analyte concentration or content.  If there is a linear relationship, test results should be evaluated by appropriate statistical methods, for example, by calculation of a regression line by the method of least squares.  In some cases, to obtain linearity between assays and sample concentrations, the test data may need to be subjected to a mathematical transformation prior to the regression analysis. Data from the regression line itself may be helpful to provide mathematical estimates of the degree of linearity.  For the establishment of linearity, a minimum of 5 concentrations is recommended.
  • 20. Example  Seven solutions containing different concentrations (0.280 – 0.520) mg/ml of ketotifen fumarate in tablets were assayed using HPLC  The results were evaluated statistically and the results shown on the following slide
  • 21. Example (continued) Concentration of ketotifen fumarate Area detected Acceptance mg/ml % criteria 0.280 70 1473566 0.320 80 1677013 0.360 90 1904848 0.400 100 2091215 0.440 110 2293647 0.480 120 2518976 0.520 130 2670144 Regression: y = ax + b 0.998 – 1.002 a = 5055766.964 b = 67608.786 r2 = 0.9984
  • 22. RANGE  The specified range is normally derived from linearity studies and depends on the intended application of the procedure.  It is established by confirming that the analytical procedure provides an acceptable degree of linearity, accuracy and precision when applied to samples containing amounts of analyte within or at the extremes of the specified range of the analytical procedure.
  • 23. Minimum Specified Ranges  for the assay of a drug substance or a finished (drug) product: normally from 80 - 120 % of the test concentration  for content uniformity, covering a minimum of 70 - 130 % of the test concentration  for dissolution testing: +/-20 % over the specified range;  e.g., if the specifications for a controlled released product cover a region from 20%, after 1 hour, up to 90%, after 24 hours, the validated range would be 0- 110% of the label claim
  • 24. Accuracy vs precision What you would like to see!
  • 25. Accuracy vs precision  Poor accuracy  Good precision
  • 26. Accuracy vs precision  Poor precision  Good accuracy
  • 27. Accuracy vs precision What would you call this?  Totally hopeless!  Poor precision  Poor accuracy
  • 28. So what definitions do these concepts lead us to in the context of assay validation?
  • 29. ACCURACY (1)  The accuracy of an analytical procedure expresses the closeness of agreement between the value which is accepted either as a conventional true value or an accepted reference value and the value found. This is sometimes termed trueness.
  • 30. ACCURACY (2) Assay of Drug Product: a) Application of the analytical procedure to synthetic mixtures of the drug product components to which known quantities of the drug substance to be analysed have been added (standard addition method); b) In cases where it is impossible to obtain samples of all drug product components, it may be acceptable either to:  add known quantities of the analyte to the drug product or  to compare the results obtained from a second, well characterized procedure, the accuracy of which is stated and/or defined (independent procedure) c) Accuracy may be inferred once precision, linearity and specificity have been established.
  • 31. ACCURACY (3) Impurities (Quantitation):  Accuracy should be assessed on samples (drug substance/drug product) spiked with known amounts of impurities.  In cases where it is impossible to obtain samples of certain impurities and/or degradation products, it is considered acceptable to compare results obtained by an independent procedure. The response factor of the drug substance can be used.  It should be clear how the individual or total impurities are to be determined e.g., weight/weight or area percent, in all cases with respect to the major analyte.
  • 32. Recommended Data  Accuracy should be assessed using a min. of 9 determinations over a min. of 3 concentration levels covering the specified range (e.g. 3 concentrations/3 replicates each of the total analytical procedure).  Accuracy should be reported as:  % recovery by the assay of known added amount of analyte in the sample or as  the difference between the mean and the accepted true value together with the confidence intervals
  • 33. Example:  Nine solutions containing different concentrations of ketotifen fumarate reference standard added to ketotifen tablet were assayed
  • 34. Example (continued): Conc. of ketotifen fumarate Area Recovery Acceptance mg/ml % detected (%) Criteria 0.280 70 1473566 99.32 0.320 80 1677013 99.48 0.360 90 1904848 100.94 0.380 95 1905862 100.51 0.400 100 2091215 100.06 0.420 105 2180374 100.03 0.440 110 2293647 100.07 0.480 120 2518976 101.01 0.520 130 2670144 98.99 Mean (recovery) : 100.04 98.0–102.0 % Standard deviation : 0.699 Relative standard deviation (RSD) : 0.699 % <2%
  • 35. PRECISION  The precision of an analytical procedure expresses the closeness of agreement (degree of scatter) between a series of measurements obtained from multiple sampling of the same homogeneous sample under the prescribed conditions.  Precision may be considered at three levels:  repeatability,  intermediate precision and  reproducibility.  Precision should be investigated using homogeneous, authentic samples. However, if it is not possible to obtain a homogeneous sample it may be investigated using artificially prepared samples or a sample solution.  The precision of an analytical procedure is usually expressed as the variance, standard deviation or coefficient of variation of a series of measurements.
  • 36. Repeatability (1)  Repeatability expresses the precision under the same operating conditions over a short interval of time.  Repeatability is also termed intra-assay precision.
  • 37. Repeatability (2)  Repeatability should be assessed using: a) a minimum of 9 determinations covering the specified range for the procedure (e.g. 3 concentrations/3 replicates each) or b) a minimum of 6 determinations at 100% of the test concentration.
  • 38. Intermediate precision  Intermediate precision expresses within-laboratories variations: different days, different analysts, different equipment, etc.  The extent to which intermediate precision should be established depends on the circumstances under which the procedure is intended to be used.  The applicant should establish the effects of random events on the precision of the analytical procedure.  Typical variations to be studied include days, analysts, equipment, etc. It is not considered necessary to study these effects individually. The use of an experimental design (matrix) is encouraged.
  • 39. Reproducibility  Reproducibility is assessed by means of an inter-laboratory trial.  Reproducibility should be considered in case of the standardization of an analytical procedure, for instance, for inclusion of procedures in pharmacopoeias.  These data are not part of the marketing authorization dossier.
  • 40. Recommended Data  The standard deviation, relative standard deviation (coefficient of variation) and confidence interval should be reported for each type of precision investigated.
  • 41. Example  The active ingredient, ketotifen fumarate, in tablets was assayed seven times using HPLC and the reference standard
  • 42. Example (continued) Sample no. Concentration (mg/ml) Area detected 1 0.4 1902803 2 0.4 1928083 3 0.4 1911457 4 0.4 1915897 5 0.4 1913312 6 0.4 1897702 7 0.4 1907019 Mean : 1910896 Standard deviation : 9841.78 Relative standard deviation (RSD) : 0.515 % Acceptance criteria: Relative standard deviation (RSD): not more than 2 %
  • 43. Detection limit vs Quantitation limit ‘Know that it’s there’ vs ‘Know how much is there’
  • 44. Detection limit (means) Is any of it present? Is it there?
  • 45. Quantitation limit How much of it is present??? How much of it is there?
  • 46. DETECTION LIMIT  The detection limit of an individual analytical procedure is the lowest amount of analyte in a sample which can be detected but not necessarily quantitated as an exact value  Several approaches for determining the detection limit are possible, depending on whether the procedure is a non- instrumental or instrumental.
  • 47. Based on Visual Evaluation  Visual evaluation may be used for non- instrumental methods but may also be used with instrumental methods.  The detection limit is determined by the analysis of samples with known concentrations of analyte and by establishing the minimum level at which the analyte can be reliably detected .
  • 48. Based on Signal-to-Noise  This approach can only be applied to analytical procedures which exhibit baseline noise.  Determination of the signal-to-noise ratio is performed by comparing measured signals from samples with known low concentrations of analyte with those of blank samples and establishing the minimum concentration at which the analyte can be reliably detected.  A signal-to-noise ratio between 3:1 or 2:1 is generally considered acceptable for estimating the detection limit.
  • 49. Based on the Standard Deviation of the Response and the Slope The detection limit (DL) may be expressed as: DL = 3.3 σ /S where σ = the standard deviation of the response S = the slope of the calibration curve The slope S may be estimated from the calibration curve of the analyte.
  • 50. Estimate of σ  Based on the Standard Deviation of the Blank  Measurement of the magnitude of analytical background response is performed by analyzing an appropriate number of blank samples and calculating the standard deviation of these responses  Based on the Calibration Curve  A specific calibration curve should be studied using samples containing an analyte in the range of DL.  The residual standard deviation of a regression line or the standard deviation of y-intercepts of regression lines may be used as the standard deviation.
  • 51. Recommended Data  The detection limit and the method used for determining the detection limit should be presented.  If DL is determined based on visual evaluation or based on signal to noise ratio, the presentation of the relevant chromatograms is considered acceptable for justification.  In cases where an estimated value for the detection limit is obtained by calculation or extrapolation, this estimate may subsequently be validated by the independent analysis of a suitable number of samples known to be near or prepared at the detection limit
  • 52. QUANTITATION LIMIT  The quantitation limit of an individual analytical procedure is the lowest amount of analyte in a sample which can be quantitatively determined with suitable precision and accuracy.  The quantitation limit is a parameter of quantitative assays for low levels of compounds in sample matrices, and is used particularly for the determination of impurities and/or degradation products.  Several approaches for determining the quantitation limit are possible, depending on whether the procedure is a non-instrumental or instrumental.
  • 53. Based on Visual Evaluation  Visual evaluation may be used for non- instrumental methods but may also be used with instrumental methods.  The quantitation limit is generally determined by the analysis of samples with known concentrations of analyte and by establishing the minimum level at which the analyte can be quantified with acceptable accuracy and precision.
  • 54. Based on Signal-to-Noise Approach  This approach can only be applied to analytical procedures that exhibit baseline noise.  Determination of the signal-to-noise ratio is performed by comparing measured signals from samples with known low concentrations of analyte with those of blank samples and by establishing the minimum concentration at which the analyte can be reliably quantified.  A typical signal-to-noise ratio is 10:1.
  • 55. Based on the Standard Deviation of the Response and the Slope  The quantitation limit (QL) may be expressed as: QL = 10 σ /S where σ = the standard deviation of the response S = the slope of the calibration curve  The slope S may be estimated from the calibration curve of the analyte.
  • 56. Estimate of σ  Based on Standard Deviation of the Blank  Measurement of the magnitude of analytical background response is performed by analyzing an appropriate number of blank samples and calculating the standard deviation of these responses.  Based on the Calibration Curve  A specific calibration curve should be studied using samples, containing an analyte in the range of QL.  The residual standard deviation of a regression line or the standard deviation of y-intercepts of regression lines may be used as the standard deviation.
  • 57. Recommended Data  The quantitation limit and the method used for determining the quantitation limit should be presented.  The limit should be subsequently validated by the analysis of a suitable number of samples known to be near or prepared at the quantitation limit.
  • 58. Robustness Small changes do not affect the parameters of the assay
  • 59. ROBUSTNESS  The robustness of an analytical procedure is a measure of its capacity to remain unaffected by small, but deliberate variations in method parameters and provides an indication of its reliability during normal usage.  The evaluation of robustness should be considered during the development phase and depends on the type of procedure under study.  If measurements are susceptible to variations in analytical conditions, the analytical conditions should be suitably controlled or a precautionary statement should be included in the procedure.  One consequence of the evaluation of robustness should be that a series of system suitability parameters (e.g., resolution test) is established to ensure that the validity of the analytical procedure is maintained whenever used.
  • 60. Typical Variations  stability of analytical solutions,  extraction time Liquid chromatography:  influence of variations of pH in a mobile phase,  influence of variations in mobile phase composition,  different columns (different lots and/or suppliers),  temperature,  flow rate. Gas chromatography:  different columns (different lots and/or suppliers),  temperature,  flow rate.
  • 61. SYSTEM SUITABILITY TESTING  System suitability testing is an integral part of many analytical procedures.  The tests are based on the concept that the equipment, electronics, analytical operations and samples to be analyzed constitute an integral system that can be evaluated as such.  System suitability test parameters to be established for a particular procedure depend on the type of procedure being validated. They are especially important in the case of chromatographic methods.
  • 62. System Suitability in Chromatography  To verify that the resolution and reproducibility of the chromatographic system are adequate for the analysis to be done  The resolution, R, is specified to ensure that closely eluting compounds are resolved from each other  Replicate injections of a standard preparation are compared to ascertain whether requirements for precision are met  The tailing factor, T, has to meet a certain requirement, because as peak asymmetry increases, integration, and hence precision, becomes less reliable
  • 63. Evaluating validation data for an HPLC procedure  Here are some suggestions………  But please note! - The slides that follow do not represent requirements; they are suggestions. - There is more than one way to do this! - Use judgement.  If you are unsure, consult with experienced analysts!!
  • 64. Specificity (selectivity) Use some or all of these procedures: - Add a synthetic mixture of excipients to the sample & check whether the assay result for the drug is the same - Add some known impurities to the test sample & check whether they are resolved (separated from) the drug - Forcably degrade the active & test whether degradants are separated from the intact drug - Assess peak purity by diode array
  • 65. Linearity - Minimum of 5 concentrations - r2 >0.99 if possible - Intercept NMT ±2% of response of 100% the working concentration - Confirm accuracy & precision over the required range
  • 66. Accuracy - Generally within +2% - Recoveries after spiking, or - Comparison with ‘well-established’ methods & by inference - Arguably can be up to +10% for related substances - What is known about the reference standard?
  • 67. Precision - repeatability  System repeatability %CV (of detector response) <2.0% for 6 injections  Method repeatability %CV <2.0% and accuracy should be within 2%
  • 68. Precision - intermediate [= ruggedness USP] - Use same complete analytical procedure for comparisons - Compare results across different analysts, days, equipment - Means preferably within 2% - Compare %CV with that for method repeatability
  • 69. Precision - reproducibility - This is not normally a component of a dossier for an application to register, but if you do have to evaluate these data then…… - For interlab comparisons - Means should preferably be within 2% - Compare the %CV with that for method repeatability - Can use an F test, normally with 95% confidence
  • 70. Limit of detection - Use some or all of these procedures: - - Visual evaluation: A clear & symmetrical peak is visible - Signal to noise ratio of 3:1 or 2:1 - Based on statistical information: - Detection limit = - 3.3 x (std dev at that concentration) - slope
  • 71. Limit of quantitation - Use some or all of these procedures: - ‘Visual’ evaluation: A clear & symmetrical peak is visible - Signal to noise ratio of 10:1 - Based on statistical information: - Detection limit = - 10 x (std dev at that concentration) - slope
  • 72. Robustness - Use some or all of these procedures: - Compare results after altering HPLC parameters, eg mobile phase composition, buffer composition, pH, column type, flow rate: - NMT ± 2% difference in assay - Compare results after storage of test solution, eg for 24h at say 250C - NMT ± 2% difference in assay
  • 73. Evaluation of analytical validation data The objective of the analytical procedure The analytical technique Item Data provided by applicant Acceptable or not? (add comments if (very briefly) necessary, & reasons if unacceptable) Is a chromatogram, spectrum or similar provided? S Linearity Range Accuracy Precision: Repeatability Precision: Intermediate Precision: Reproducibility Detection limit Quantitation limit Robustness System suitability (if necessary) Are the data concerning analytical validation satisfactory? YES/NO DataIfon therecommended questions to the applicant appear in NO, reference standard …………………………………………………………………… Other evaluator comments: (eg page number below, or draft letter to the company on page……)

Notas do Editor

  1. ‘ -’ signifies that this characteristic is not normally evaluated ‘ +’ signifies that this characteristic is normally evaluated (1) In cases where reproducibility has been performed . Intermediate precision is not needed. (2) Lack of specificity of one analytical procedure could be compensated by other supporting analytical procedures (3) May be needed in some cases.
  2. NB From perspective of an evalr, not from perspective of an analyst experienced in HPLC.
  3. Selectivity: Provide representative chromatograms with labelled peaks Suitable acceptance criteria might be: Capacity factor k’ &gt; 1.0 Resolution &gt;1.0 If ref imps not available, can prepare a suitable solution by forced degradation. If degradation &gt;10%, may have to argue relevance. Diode array (suggested by ICH) may not be sensitive to low levels of imps, esp if chromophores similar When validating ID tests, remember to consider other actives on your site, as well as excipients
  4. Linearity: Normally submit plot of detector response vs concentration, plus regression analysis Normally also determine accuracy and precision at extremes of range TGAL usually test across range of 50-150%, especially when test is to be used for CU and/or dissolution testing
  5. Accuracy: When need to determine accuracy over a range of concentrations, normally 3 concentrations sufficient If mixture of excipients not available, may add known quantities of active and determine difference in results Care with reference substance. If a working standard (ie qualified wrt pharmacopoeial ref subst), bear in mind possibility that errors can accumulate. Care with hydration. Watch for time dependent variability, possibly associated with decomposition. If sample is filtered during preparation, specify type of filter and check for losses (soln of ref filtered vs not filtered)
  6. ‘ System’ repeatability: Aka repeatability between injections %CV can be of the order of 0.2%, particularly when automated FDA recommends min of 10 injections with %CV &lt; 1.0% Many EP monographs prescribe %CV &lt; 1% Acceptable %CV depends in part on the acceptance range for the assay in question Method repeatability: Aka repeatability of the complete method In practice, often estimated in conjunction with accuracy Means should be within + 2% of t/c Typically measured by: 6 replicates at 100% t/c OR 3 replicates of 3 concentrations %CV can be of the order of 0.4%, particularly when automated Acceptable %CV may be higher for: Impurities More complex matrices (such as creams) Microdose products
  7. Intermediate precision: FDA uses the term ‘ruggedness’ with same meaning Although slide states means preferably within 2%, criteria are not laid down Acceptability is assessed on case by case basis
  8. Reproducibility: Again: although slide states means preferably within 2%, criteria are not laid down Again: acceptability is assessed on case by case basis
  9. LOD: LOD especially important for imps Submit typical chromatograms at LOD Std dev may be estimated from: Std dev of blank samples Std dev of intercept Residual std dev from regression analysis of calibration plot
  10. LOQ: Again important for imps LOQ must be &lt; acceptance criterion for the imp(s) !! Personally I would not want to rely on ‘visual’ evaluation for limit of quantitation. Submit typical chromatograms at LOQ As for LOD, std dev may be estimated from: Std dev of blank samples Std dev of intercept Residual std dev from regression analysis of calibration plot Confirm the LOQ by determining %CV at that value
  11. Dot point 1: - If difference in assay &gt;2%, describe attempts to improve. If little improvement gained, insert warnings into write-up of method. Dot point 2: - Duration and conditions of storage should represent most stressful situation likely to occur, eg during a 24hour auto-sampling run.