SlideShare a Scribd company logo
ANALYTICAL METHOD (DEVELOPMENT,
VALIDATION, TRANSFER & LIFE CYCLE
MANAGEMENT)
Prepared by: MD. MIZANUR RAHMAN MIAJEE
1
ANALYTICAL METHOD VALIDATION
REGULATORY GUIDANCE:
• ICH Q2B- Validation of Analytical Procedures: Methodology
• USP <1225> Validation of Compendial Procedures
• USP <1226> Verification of Compendial Procedures
• USP <1220> The Analytical Procedure Lifecycle – Proposed
• USP <621> Chromatography
• FDA Guidance- Analytical Procedures and Methods Validation for Drugs
and Biologics
• CDER Reviewer Guidance- Validation of Chromatographic Methods
• Other Journal
2
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Principle/Scope: A description of the basic principles of the
analytical test/technology (i.e., separation, detection); target
analyte(s) and sample(s) type (e.g., drug substance, drug
product, impurities)
• Apparatus/ Equipment: All required qualified equipment and
components (e.g., instrument type, detector, column type,
dimensions, and alternative column, filter type).
3
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Operating Parameters: Qualified optimal settings and ranges
(include allowed adjustments supported by compendial
sources or development and/or validation studies) critical to
the analysis (e.g., flow rate, components temperatures, run
time, detector settings, gradient, head space sampler). A
drawing with experimental configuration and integration
parameters may be used, as applicable.
4
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
Reagents/Standards: The following should be listed where
applicable:
• Description of reagent or standard
• Grade of chemical (e.g., USP/NF, American Chemical Society,
High Performance or Pressure Liquid Chromatography, or
Gas Chromatography and preservative-free)
• Source (e.g., USP reference standard, qualified in-house
reference material, WHO International Standard/Reference
Material, CBER standard)
5
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Purity (for pure chemicals only), State (e.g., dried, undried),
and concentration
• Potencies (where required by CFR, USP)
• Storage conditions
• Directions for safe use (as per current Safety Data Sheet)
• Validated or documented shelf life
6
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
Sample Preparation:
Procedures (e.g., extraction method, dilution or
concentration, desalting procedures and mixing by sonication,
shaking or sonication time) for the preparations for individual
sample tests. A single preparation for qualitative and
replicate preparations for quantitative tests with appropriate
units of concentrations for working solutions (e.g., µg/ml or
mg/ml) and information on stability of solutions and storage
conditions.
7
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Standards Control Solution Preparation: Procedures for the
preparation and use of all standard and control solutions with
appropriate units of concentration and information on
stability of standards and storage conditions, including
calibration standards, internal standards, system suitability
standards, etc.
8
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Procedure: A step-by-step description of the method (e.g.,
equilibration times, and scan/injection sequence with blanks,
placeboes, samples, controls, sensitivity solution (for impurity
method) and standards to maintain validity of the system
suitability during the span of analysis) and allowable
operating ranges and adjustments if applicable.
9
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• System Suitability: Confirmatory test(s) procedures and
parameters to ensure that the system (equipment,
electronics, and analytical operations and controls to be
analyzed) will function correctly as an integrated system at
the time of use. The system suitability acceptance criteria
applied to standards controls and samples, such as peak
tailing, precision and resolution acceptance criteria, may be
required as applicable.
10
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Calculations: The integration method and representative
calculation formulas for data analysis (standards, controls,
samples) for tests based on label claim and specification (e.g.,
assay, specified and unspecified impurities and relative
response factors). This includes a description of any
mathematical transformations or formulas used in data
analysis, along with a scientific justification for any correction
factors used.
11
ANALYTICAL METHOD VALIDATION
CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA):
• Data Reporting: A presentation of numeric data that is consistent with
instrumental capabilities and acceptance criteria. The method should
indicate what format to use to report results (e.g., percentage label claim,
weight/weight, and weight/volume) with the specific number of
significant figures needed. The American Society for Testing and Materials
(ASTM) E29 standard describes a standard practice for using significant
digits in test data to determine conformance with specifications. For
chromatographic methods, you should include retention times (RTs) for
identification with reference standard comparison basis, relative retention
times (RRTs) (known and unknown impurities) acceptable ranges and
sample results reporting criteria
12
ANALYTICAL METHOD VALIDATION
GENERAL POINT MUST BE CONSEDERED FOR ANALYTICAL METHOD
(FDA Reviewer Guidance):
• The sample and standard should be dissolved in the mobile
phase. If that is not possible, then avoid using too high level
of the organic solvent as compared to the level in the mobile
phase.
• The sample and standard concentrations should be close if
not the same.
• The samples should be bracketed by standards during the
analytical procedure
13
ANALYTICAL METHOD VALIDATION
GENERAL POINT CONSEDER FOR ANALYTICAL METHOD:
• Filtration of the samples before injection is occasionally
observed. Filtration will remove particulates (centrifugation
performs the same function) that may clog columns.
Adhesion of the analyte to the filter can also happen. This will
be of importance especially for low level impurities. Data to
validate this aspect should be submitted by the applicant.
14
ANALYTICAL METHOD VALIDATION
15
ANALYTICAL METHOD VALIDATION
• An analytical procedure is developed to test a defined characteristic
of the drug substance or drug product against established
acceptance criteria for that characteristic.
• Method development should begin with an initial risk assessment
and follow with multivariate experiments. Knowledge gained during
these studies on the sources of method variation can help you
assess the method performance.
ANALYTICAL METHOD DEVELOPMENT
16
ANALYTICAL METHOD VALIDATION
What is QbD
A systematic approach to development that begins with
predefined objectives and emphasizes product and process
understanding and process control, based on sound science
and quality risk management.
Benefits of Application of QbD Approach to Analytical Methods
• Development of a robust method
• Applicable throughout the life cycle of the product
Regulatory flexibility −
• Movements within “Design Space” are not considered a change in
method.
ANALYTICAL METHOD DEVELOPMENT
17
ANALYTICAL METHOD VALIDATION
ANALYTICAL METHOD DEVELOPMENT
18
ANALYTICAL METHOD VALIDATION
• What Is An ATP?
• Analytical Target Profile is a part of QbD which is a predefined,
documented statement of the requirements for the quality of the
reportable value, which the analytical procedure must generate in
order for the procedure to be considered fit for purpose Using
Current Industry Acceptance Criteria. The procedure must be able
to accurately quantify [drug] in the [FPP] in the presence of
[Impurity/degradation product] with the following requirements for
the reportable values: Accuracy = 100.0% ± 3.0% and precision ≤
2.0% relative standard deviation (RSD).
ANALYTICAL METHOD DEVELOPMENT
19
ANALYTICAL METHOD VALIDATION
• What Is in ATP?
In ATP Target profile for the analytical method to be predefined. Some
example are given below:
1. Sample sonication time: as per QbD different sonicatione time
should be considered to get optimum solubility (ATP)
2. Chromatographic Run Time: A target run time should be selected
and as per QbD, different run time to be performed to get optimum
one.
3. Sample & standard solution concentration in impurity analysis:
Concentration of sample is very important in impurity analysis to
cover Reporting threshold limit as per ICH Q2B. A target
concentration to be selected before development work.
ANALYTICAL METHOD DEVELOPMENT
20
ANALYTICAL METHOD VALIDATION
Parameters that may be evaluated during method development:
During early stages of method development, the robustness of methods
should be evaluated because this characteristic can help you decide which
method you will submit for approval. Other characteristics that may be
considered during method development are:
• Specificity
• Linearity
• Accuracy
• Precision
• Range
• Quantitation limit
• Detection limit.
• Solution Stability
ANALYTICAL METHOD DEVELOPMENT
21
ANALYTICAL METHOD VALIDATION
ANALYTICAL METHOD DEVELOPMENT
Describe the
sample
Establish
Goals
Consider sample
preparation
Choose
detector
Choose
chromatographic
mode
First Run
Optimize
separation
Predict
Problems
Validate and
Release
method
22
ANALYTICAL METHOD VALIDATION
Collection of literature:
Analytical method development starts with literature search
where in various pharmacopeias like USP, EP, BP, JP etc and
chromatographic journals are referred to check the
availability of suitable analytical methods. If any suitable
method is found, it is still necessary to perform method
optimization and validation to prove that the method can be
successfully adapted for its intended use.
23
ANALYTICAL METHOD VALIDATION
Sample information: The synthetic route from raw material to
finished dosage forms (structures) of the molecule should be
collected and the impurities originating from starting materials
of synthesis, degradation products, excipients, solvents etc.
need to be considered. The method should be designed
based on the closely related structures and get the best
resolution between the closely related compounds. The
structures of impurities, starting material, intermediates and
degradation products are compared with the structure of drug
substances and arrive at the polarity whether they are less
polar or more polar than the compound of interest.
24
ANALYTICAL METHOD VALIDATION
• Selection of diluents: The diluent should be chosen based on
the solubility of impurities, degradation products, starting
materials, intermediates and the analyte. The diluent should
be compatible with the mobile phase to get a better peak
shape of analyte.
25
ANALYTICAL METHOD VALIDATION
• Selection of stationary phase: The selection of bonded phase can
be based on the polarity of molecule and its by-products. For RP-LC,
a wide variety of columns are accessible covering a large range of
polarity by cross-linking Si-OH groups with linear alkyl chains like
C8, C18 and phenyl groups (-C6H6), nitrile groups (-CN), , different
embedded hybridized groups amino groups (-NH2) etc. C18
columns are the commonly used columns in HPLC method analysis.
C8 or Octyl bonded phases are also used occasionally. Like C18,
they are non-polar, but not as hydrophobic.
26
ANALYTICAL METHOD VALIDATION
• Detectors:
Various detectors used in HPLC instrument include UV-Visible
detector, photodiode array detector, fluorescence detector,
conductivity detector, refractive index detector, electro-
chemical detector, mass spectrometer detector and
evaporative light scattering detector. UV-Visible detectors are
typical in many laboratories as they can detect a wide array of
compounds
27
ANALYTICAL METHOD VALIDATION
• pH range:
Method development within the different pH ranges from 1 to
12 for better chromatographic resolution between two or
more peaks of an analyte depends upon three main factors,
column efficiency, selectivity and retention time. The
ionizable analytes are either bases or acids and they affect
the above three factors dramatically with change in pH.
Retention time can be improved by changing the pH that will
lead to easy separation of ionizable analytes from non-ionized
forms.
28
ANALYTICAL METHOD VALIDATION
• pH range:
To achieve optimum resolution, it requires change in the pH of
mobile phase. Method development can proceed by
investigating parameters of chromatographic separations first
at low pH and then at higher pH until optimum results are
achievd.
29
ANALYTICAL METHOD VALIDATION
• Mobile phase composition:
Mobile phase composition (or solvent strength) plays an
important role in RP-HPLC separation. Acetonitrile (ACN),
methanol (MeOH) and tetrahydrofuran (THF) are commonly
used solvents in RP -HPLC having low UV cut- off of 190, 205
and 212nm respectively. These solvents are miscible with
water. Mixture of acetonitrile and water is the best initial
choice for the mobile phase during method development.
30
ANALYTICAL METHOD VALIDATION
• Column temperature:
Separation of many samples can be enhanced by selecting the
right column temperature. Higher column temperature
reduces system backpressure by decreasing mobile phase
viscosity, which in turn allows use of longer columns with
higher separation efficiency. The optimum temperature is
dependent upon the nature of the mixture components. The
overall separation can be improved by the simultaneous
changes in column temperature and mobile phase
composition
31
ANALYTICAL METHOD VALIDATION
• Analytical Method Transfer:
What is analytical method transfer: It is the documented
process that qualifies a laboratory (the receiving unit) to use
an analytical test procedure that originated in another
laboratory (the transferring unit), thus ensuring that the
receiving unit has the procedural knowledge and ability to
perform the transferred analytical procedure as intended.
32
ANALYTICAL METHOD VALIDATION
• Transfer may be-
• One R & D development lab to another R & D development
lab
• R & D development lab to quality control lab inside same unit
• R & D development lab to quality control lab of other unit
• One facility to another intra-company facility
• One company to another company such as a contract
research organization
33
ANALYTICAL METHOD VALIDATION
Procedure of Analytical Method Transfer: Analytical
Method Transfer (AMT) can be done by two ways:
1. Comparative Testing
2. Co-validation between Two or More Laboratories
34
ANALYTICAL METHOD VALIDATION
1. Comparative Testing: analysis of a predetermined number of
samples of the same lot by both the sending and the receiving
units.
Prerequisite: A preapproved transfer protocol prepared by
sending unit that stipulates the details of the procedure, the
samples that will be used, and the predetermined acceptance
criteria, including acceptable variability
Criteria: Meeting the predetermined acceptance criteria is
necessary to assure that the receiving unit is qualified to run
the procedure.
35
ANALYTICAL METHOD VALIDATION
2. Co-validation Between Two or More Laboratories: The
transferring unit can involve the receiving unit in an inter
laboratory co-validation, including them as a part of the
validation team at the transferring unit and thereby obtaining
data for the assessment of reproducibility
Prerequisite: a preapproved transfer or validation protocol
that provides the details of the procedure, the samples to be
used, and the predetermined acceptance criteria
Criteria: Meeting the predetermined acceptance criteria is
necessary to assure that the receiving unit is qualified to run
the procedure.
36
ANALYTICAL METHOD VALIDATION
Transfer Waiver:
• The new product’s composition is comparable to that of an
existing product and/or the concentration of active ingredient
is similar to that of an existing product and is analyzed by
procedures with which the receiving unit already has
experience
• The analytical procedure being transferred is described in the
USP–NF, and is unchanged. Verification should apply in this
case
• The analytical procedure transferred is the same as or very
similar to a procedure already in use.
37
ANALYTICAL METHOD VALIDATION
Transfer Waiver:
• The personnel in charge of the development, validation, or
routine analysis of the product at the transferring unit are
moved to the receiving unit. If eligible for transfer waiver, the
receiving unit should document it with appropriate
justifications.
38
ANALYTICAL METHOD VALIDATION
• ELEMENTS RECOMMENDED FOR THE TRANSFER OF
ANAYTICAL PROCEDURES:
• Transferring unit:
• provide training to the receiving unit, training should be
documented, providing the analytical procedure, the
reference standards, the validation reports, and any
necessary documents, as well as for providing the necessary
training and assistance to the receiving unit as needed during
the transfer.
39
ANALYTICAL METHOD VALIDATION
• ELEMENTS RECOMMENDED FOR THE TRANSFER OF
ANAYTICAL PROCEDURES:
• Receiving unit:
• Provides qualified staff or properly trains the staff before the
transfer, ensures that the facilities and instrumentation are
properly calibrated and qualified as needed, and verifies that
the laboratory systems are in compliance with applicable
regulations and in-house general laboratory procedures.
40
ANALYTICAL METHOD VALIDATION
• ELEMENTS RECOMMENDED FOR THE TRANSFER OF
ANAYTICAL PROCEDURES:
• Both unit: Compare and discuss data as well as any deviations
from the protocol. This discussion addresses any necessary
corrections or updates to the final report and the analytical
procedure as necessary to reproduce the procedure. A single
lot of the article
41
ANALYTICAL METHOD VALIDATION
• ELEMENTS RECOMMENDED FOR THE TRANSFER OF
ANAYTICAL PROCEDURES:
• Content of transfer protocol: Below mentioned parameters
must be included in transfer protocol
• Objective
• Scope
• Responsibilities of the transferring and receiving units,
• Materials and instruments that will be used,
• Analytical procedure
• Experimental design
• Acceptance criteria for all tests and/or methods included in
the transfer. 42
ANALYTICAL METHOD VALIDATION
• Analytical Procedure for AMT: The procedure should be
written with sufficient detail and explicit instructions, so that
a trained analyst can perform it without difficulty. A pre-
transfer meeting between the transferring and receiving units
is helpful to clarify any issues and answer any questions
regarding the transfer process. If complete or partial
validation data exist, they should be available to the receiving
unit, along with any technical details required to perform the
test in question.
43
ANALYTICAL METHOD VALIDATION
• What is analytical method validation
Validation is the process of demonstrating that an analytical
procedure is suitable for its intended purpose
44
ANALYTICAL METHOD VALIDATION
• Importance of Analytical Method Validation
 Validation of a method involves using experimental design to prove that the
method can produce accurate and precise results within the scope of its
intended use
 Understanding the application and limitations of the test method will allow for
accurate assessment of sample information, ranging from process outputs to
commercial release testing and many steps in between.
 If the method validation has not been performed or has been performed in an
inadequate manner, the method is not proven to provide reliable data.
• A majority of audit findings fell into three main categories: 1. the use of a non-
validated method for critical decision making; 2. inadequate method
validation that did not provide the necessary information; 3. method
validation that lacked appropriate controls to maintain the integrity of the
validation 45
ANALYTICAL METHOD VALIDATION
• Types of Analytical Method need to validate
 Identification tests
 Quantitative tests for impurities;
 Limit tests for the control of impurities; and
 Assay tests for the active component in a drug substance, drug product,
or other selected component(s) in the drug product.
46
ANALYTICAL METHOD VALIDATION
• Noncompendial Analytical Procedures: Requires Complete Validation
• Compendial Analytical Procedures: Requires Verification
47
ANALYTICAL METHOD VALIDATION
• Why verification:
 Drug substance's synthetic route,
 The method of manufacture for the drug product
 Drug substances from different suppliers may have different impurity
profiles that are not addressed by the compendial test procedure
 The excipients in a drug product can vary widely among manufacturers
and may have the potential to directly interfere with the procedure or
cause the formation of impurities that are not addressed by the
compendia procedure
 Drug products containing different excipients, antioxidants, buffers, or
container extractives may affect the recovery of the drug substance from
the matrix
(But as per current thinking of FDA, FDA prefer complete validation for
compendia method instead of verification)
48
ANALYTICAL METHOD VALIDATION
• Following parameter to be considered for Analytical Method
Validation:
 Specificity
 Accuracy
 Precision (Repeatability, Intermediate Precision & Reproducibility)
 Linearity
 Range
 Quantitation limit (LOQ)
 Detection limit (LOD)
 Robustness
49
ANALYTICAL METHOD VALIDATION
SPECIFICITY /SELECTIVITY
Ability to assess unequivocally the analyte in the presence of components
that may be expected to be present, such as impurities, degradation
products, and matrix components.
• Identification: Ensure the identity of the analyte.
• Impurity: Ensure that all of the analytical procedures performed allow an
accurate statement of the content of impurities of an analyte (e.g.,
related substances test, heavy metals limit, or organic volatile impurities).
• Assays: Provide an exact result, which allows an accurate statement on
the content or potency of the analyte in a sample
50
ANALYTICAL METHOD VALIDATION
SPECIFICITY /SELECTIVITY
• Procedure:
When Impurities are available (Specified identified impurity): 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 (by
comparison with the assay result obtained on unspiked samples). For the
impurity test, the discrimination may be established by spiking drug
substance or drug product with appropriate levels of impurities and
demonstrating the separation of these impurities individually and/or from
other components in the sample matrix.
51
ANALYTICAL METHOD VALIDATION
Accuracy
The accuracy of an analytical procedure is the closeness of test results
obtained by that procedure to the true value.
Evaluate by analyzing synthetic mixtures: known quantity of API with
excipients/ Impurity
• Recommended Data: Accuracy should be assessed using a minimum of 9
determinations over a minimum 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 percent 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.
52
ANALYTICAL METHOD VALIDATION
Precision
The precision of an analytical procedure is the degree of agreement among
individual test results when the procedure is applied repeatedly to
multiple samplings of a homogeneous sample. The precision of an
analytical procedure is usually expressed as the standard deviation or
relative standard deviation (coefficient of variation) of a series of
measurements
A. Repeatability
• Repeatability should be assessed using:
(1) A minimum of 9 determinations covering the specified range for the
procedure (e.g., 3 concentrations/3 replicates each); or
(2) A minimum of 6 determinations at 100% of the test concentration.
53
ANALYTICAL METHOD VALIDATION
Precision
B. Intermediate Precision
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 necessary to study these effects
individually. The use of an experimental design (matrix) is encouraged.
54
ANALYTICAL METHOD VALIDATION
Precision
C. 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
55
ANALYTICAL METHOD VALIDATION
Linearity & Range
Linearity: The linearity of an analytical procedure is its ability to elicit test results that
are directly, or by a well-defined mathematical transformation, proportional to the
concentration of analyte in samples within a given range
Parameter to check: The correlation coefficient, y-intercept, slope of the regression
line, and residual sum of squares should be submitted.
Number of sample/ Injection: For the establishment of linearity, a minimum of five
concentrations is recommended. Other approaches should be justified.
56
ANALYTICAL METHOD VALIDATION
Linearity & Range
Range: The range of an analytical procedure is the interval between the upper and
lower levels of analyte (including these levels) that have been demonstrated to be
determined with a suitable level of precision, accuracy, and linearity using the
procedure as written assay of a drug substance or a finished (drug) product:
Normally from 80% to 120% of the test concentration
For content uniformity: Covering a minimum of 70% to 130% of the test
concentration, unless a wider, more appropriate range, based on the nature of the
dosage form (e.g., metered dose inhalers), is justified
57
ANALYTICAL METHOD VALIDATION
Linearity & Range
• 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
• For the determination of an impurity: From the reporting level of an impurity to
120% of the specification. For impurities known to be unusually potent or to
produce toxic or unexpected pharmacological effects, the detection/quantitation
limit should be commensurate with the level at which the impurities must be
controlled.
• If assay and purity are performed together as one test and only a 100% standard is
used, linearity should cover the range from the reporting level of the impurities to
120% of the assay specification.
58
ANALYTICAL METHOD VALIDATION
Detection limit
It is the lowest amount of analyte in a sample that can be detected, but not
necessarily quantitated, under the stated experimental conditions. Thus,
limit tests merely substantiate that the amount of analyte is above or
below a certain level. The detection limit is usually expressed as the
concentration of analyte (e.g., percentage or parts per billion) in the
sample.
A. 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 59
ANALYTICAL METHOD VALIDATION
Detection limit
B. 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.
Signal-to-noise ratio between 3 or 2:1 is generally considered acceptable for
estimating the detection limit.
60
ANALYTICAL METHOD VALIDATION
Detection limit
C. Based on the Standard Deviation of the Response and the Slope
The detection limit (DL) may be expressed as:
where
= 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. The
estimate of may be carried out in a variety of ways, for example:
61
ANALYTICAL METHOD VALIDATION
Detection limit
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.
62
ANALYTICAL METHOD VALIDATION
Detection limit
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.
63
ANALYTICAL METHOD VALIDATION
Quantitation Limit
The quantitation limit is a characteristic of quantitative assays for low levels
of compounds in sample matrices, such as impurities in bulk drug
substances and degradation products in finished pharmaceuticals. It is the
lowest amount of analyte in a sample that can be determined with
acceptable.
Precision and Accuracy under the stated experimental conditions.
The quantitation limit is expressed as the concentration of analyte (e.g.,
percentage or parts per billion) in the sample.
QL to be determined as like Detection limit except signal-to-noise ratio is
10:1. and the calculation would be 64
ANALYTICAL METHOD VALIDATION
Quantitation Limit
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
65
ANALYTICAL METHOD VALIDATION
Robustness
The evaluation of robustness should be considered during the development
phase and depends on the type of procedure under study. It should show
the reliability of an analysis with respect to deliberate variations in
method parameters.
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.
66
ANALYTICAL METHOD VALIDATION
Robustness
• Examples of typical variations are:
o Stability of analytical solutions
o Extraction time
• In the case of liquid chromatography, examples of typical variations are:
o Influence of variations of pH in a mobile phase
o Influence of variations in mobile phase composition
o Different columns (different lots and/or suppliers)
o Temperature
o Flow rate
67
ANALYTICAL METHOD VALIDATION
Robustness
System suitability testing: These tests are used to verify that the
chromatographic system is adequate for the intended analysis. system
suitability tests are performed by collecting data from replicate injections
of standard or other solutions as specified in the individual monograph.
• Factors that may affect chromatographic behavior include the following:
Composition, ionic strength, temperature, and apparent pH of the mobile
phase•
o Flow rate, column dimensions, column temperature, and pressure
o Stationary phase characteristics, including type of chromatographic support
(particle-based or monolithic), particle or macropore size, porosity, and
specific surface area
o Reverse-phase and other surface modification of the stationary phases, the
extent of chemical modification (as expressed by end-capping, carbon loading,
and others) 68
ANALYTICAL METHOD VALIDATION
Robustness
System Suitability: Unless otherwise specified in the individual monograph,
data from five replicate injections of the analyte are used to calculate the
relative standard deviation (RSD), if the requirement is ≤ 2.0%; data from
six replicate injections are used if the RSD requirement is >2.0%.
If adjustments of operating conditions are necessary in order to meet system
suitability requirements, each of the items in the following list is the
maximum variation that can be considered:
69
ANALYTICAL METHOD VALIDATION
Robustness
pH of mobile phase (HPLC): ±0.2 units of the value or range specified.
Concentration of salts in buffer (HPLC): ±10% if the permitted pH variation
Ratio of components in mobile phase (HPLC): ±30% relative. However, the
change in any component cannot exceed ±10% absolute (i.e., in relation
to the total mobile phase)
Wavelength of UV-visible detector (HPLC): ±3 nm
Stationary phase: Length- between −25% and 50% of the prescribed L/dp
Flow rate (HPLC): ±50% (Based on other factors)
Injection volume (HPLC): Can be adjusted as far as it is consistent with
accepted precision, linearity, and detection limits
Column temperature (HPLC): As much as ±10°.
70
ANALYTICAL METHOD VALIDATION
Life Cycle Management of Analytical Procedure
Once an analytical procedure (including compendial methods) is successfully
validated or verified and implemented, the procedure should be followed
during the life cycle of the product to continually assure that it remains fit
for its intended purpose.
71
ANALYTICAL METHOD VALIDATION
Life Cycle Management of Analytical Procedure
What to be done:
 Trend analysis on method performance at regular intervals
 New information and risk assessments (e.g., a better understanding of
product CQAs or awareness of a new impurity) may warrant the
development and validation of a new or alternative analytical method
 An appropriate number of retention samples should be maintained to
allow for comparative studies.
 The retention samples used in comparative studies should include
samples that represent marketed product and, when possible, pivotal
clinical trial material.
72
ANALYTICAL METHOD VALIDATION
Life Cycle Management of Analytical Procedure
Re-Validation:
 When a change is made to an analytical procedure, such as a change in
equipment or reagent, then re-validation of all or part of the analytical
procedure needs to be considered.
 Method re-validation should focus on the critical performance
characteristics of the method, such as specificity, precision, and accuracy.
 The scope of re-validation should be risk-based.
73
ANALYTICAL METHOD VALIDATION
Life Cycle Management of Analytical Procedure
• Analytical Method Comparability Studies:
Alternative Analytical Procedures
An alternative analytical procedure is an analytical procedure that is used in
place of the FDA approved analytical procedure. After approval, for an
NDA or ANDA, or for a procedure approved in a BLA but not included in an
FDA regulation, the addition, revision, or deletion of an alternative
analytical procedure that provides the same or increased assurance of the
identity, strength, quality, purity, or potency of the material being tested
as the analytical procedure described in the approved application, must
be documented in the next annual report
74
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
If a procedure is a validated quantitative analytical procedure that can detect
changes in a quality attribute(s) of the drug substance and drug product
during storage, it is considered a stability- indicating test. To demonstrate
specificity of a stability-indicating test, a combination of challenges should
be performed. Some challenges include the use of samples spiked with
target analytes and all known interferences; samples that have undergone
various laboratory stress conditions; and actual product samples
(produced by the final manufacturing process) that are either aged or
have been stored under accelerated temperature and humidity
conditions.
75
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
Objective
The main objective of a stability indicating method is to monitor results
during stability studies in order to guarantee safety, efficacy and quality. It
represents also a powerful tool when investigating out-of-trend (OOT) or
out-of-specification (OOS) results in quality control processes.
Important factor to consider: The method should be sensitive to the
reportable impurity level, LOQ (Limit of Quantitation), which is typically
0.05% of label claim, should be established in the method, and the
method should be linear from LOQ to typically up to 150% of the nominal
standard (STD) concentration
76
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
77
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
• How develop a SIM:
Generation of degraded samples for testing selectivity of the method
Determination of Limit of Quantification (LoQ) In close relation to the
determination of the amount of degradation is the evaluation of Limit of
Detection (LoD) and Limit of Quantification (LoQ) of the method. These
limits should be closely related to the Reporting, Identification and
Qualification of degradation products, as stated in ICH Q3B (R2). These
thresholds are determined either as percentage of drug substance or total
daily intake (TDI) of degradation product.
78
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
• Force Degradation:
Objective of forced degradation studies:
 To establish degradation pathways of drug substances and drug products.
 To differentiate degradation products that are related to drug products from those
that are generated from non-drug product in a formulation.
 To elucidate the structure of degradation products.
 To determine the intrinsic stability of a drug substance in formulation.
 To reveal the degradation mechanisms such as hydrolysis, oxidation, thermolysis
or photolysis of the drug substance and drug product
 To establish stability indicating nature of a developed method.
 To understand the chemical properties of drug molecules.
 To generate more stable formulations.
 To produce a degradation profile similar to that of what would be observed in a
formal stability study under ICH conditions.
• To solve stability-related problems 79
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
80
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
What to include: The testing should include the effect of temperatures (in
10°C increments (ie, 50°C, 60°C) above that for accelerated testing),
humidity (ie, 75% relative humidity or greater) where appropriate,
oxidation, and photolysis on the drug substance. The testing should also
evaluate the susceptibility of the drug substance to hydrolysis across a
wide range of pH values when in solution or suspension.”
81
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
82
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
How much is enough
Values anywhere between 5% to 20% degradation of the drug substance
have been considered as reasonable and acceptable for validation of
chromatographic assays. However, for small pharmaceutical molecules for
which acceptable stability limits of 90% of label claim is common,
pharmaceutical scientists have agreed that approximately 10%
degradation is optimal for use in analytical validation.
83
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
Selection of drug concentration
Which concentration of drug should be used for degradation study has not
been specified in regulatory guidance. It is recommended that the studies
should be initiated at a concentration of 1 mg/mL . By using drug
concentration of 1 mg/mL, it is usually possible to get even minor
decomposition products in the range of detection
84
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
What is peak purity: Peak purity is an analysis of spectral differences,
assuming the impurities are spectrally different from the analyte. The
spectral analysis is achieved using vector analysis algorithms The more
similar the spectra are, the closer the value is to 0.0 degrees; the more
spectrally different, the larger the value (90 degrees is the maximum).
When no significant spectral differences are found within a peak, one can
conclude that the peak is spectrally homogeneous and probably contains
a single compound.
85
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
 Peak Purity algorithm analyzes all spectra within a peak
 The Apex Spectrum is the reference spectrum that it compares to.
 If Purity Angle < Purity Threshold, Within the noise of the system the peak
is spectrally homogeneous or spectrally pure.
 If Purity Angle > Purity Threshold , there is something within the peak that
cannot be explained by noise. The peak is not spectrally pure, the Purity
Flag field is checked.
86
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
Purity Angle > Purity Threshold = Impure Peak
87
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
Purity threshold > Purity Angle = Pure Peak
88
ANALYTICAL METHOD VALIDATION
Stability Indicating Method (SIM)
Mass balance
• Mass balance correlates the measured loss of a parent drug to the
measured increase in the amount of degradation products. It is a good
quality control check on analytical methods to show that all degradation
products are adequately detected and do not interfere with quantitation
of the parent drug (i.e.,stability-indicating methods).
89
ANALYTICAL METHOD VALIDATION
System Suitability
General Recommendation:
System suitability testing is essential for the assurance of the quality
performance of the chromatographic system. The amount of testing
required will depend on the purpose of the test method. For dissolution
or release profile test methods using an external standard method, k', T
and RSD are minimum recommended system suitability tests. For
acceptance, release, stability, or impurities / degradation methods using
external or internal standards, k', T, R, and RSD are recommended as
minimum system suitability testing parameters.
90
ANALYTICAL METHOD VALIDATION
System Suitability
Capacity factor (k'): k' = (tR - to) /to
The capacity factor is a measure of where the peak of interest is located with
respect to the void volume, i.e., elution time of the non-retained
components.
Recommendations: The peak should be well-resolved from other peaks and
the void volume. Generally the value of k' is > 2.
Void volume: amount of dead volume in the column that is not taken up by
the particle size of the stationary phase
91
ANALYTICAL METHOD VALIDATION
System Suitability
Resolution (R,):
R, is a measure of how well two peaks are separated. For reliable
quantitation, well-separated peaks are essential for quantitation. This is a
very useful parameter if potential interference peak(s) may be of concern.
The closest potential eluting peak to the R, is minimally influenced by the
ratio of the two compounds being measured. The resolution of peaks as
indicated by the R, values is shown in below Figure:
Recommendations: R, of > 2 between the peak of interest and the closest
potential interfering peak (impurity, excipient, degradation product,
internal standard, etc.) is desirable. 92
ANALYTICAL METHOD VALIDATION
System Suitability
Tailing factor (T):
The accuracy of quantitation decreases with increase in peak tailing because
of the difficulties encountered by3the integrator in determining
wheretwhen the peal< ends and hence the calculation of the area under
the peal peak of interest. Below Figures illustrate the tailing factors and
the effect on quantitation. If the integrator is unable to determine exactly
when an upslope or downslope occurs, ' accuracy drops.
Recommendations: T of less than 2
93
ANALYTICAL METHOD VALIDATION
System Suitability
Theoretical plate number (N):
Theoretical plate number is a measure of column efficiency, that is, how
many peaks can be located per unit run-time of the chromatogram. N is
fairly constant for each peak on a chromatogram with a fixed set of
operating conditions.
Recommendations: The theoretical plate number depends on elution time
but in general. should be > 2000.
94
ANALYTICAL METHOD VALIDATION
System Suitability
Precision /injection repeatability (RSD): Injection precision expressed as RSD
(relative standard deviation) indicates the performance of the HPL
chromatograph which includes the plumbing, column, and environmental
conditions, the time the samples are analyzed. It should be noted that
sample preparation and manufacturing variations are not considered.
Recommendations: RSD of s 1% is desirable.
95
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
96
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
97
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
98
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
99
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
100
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
101
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
102
ANALYTICAL METHOD VALIDATION
Example of FDA Warning letter
103

More Related Content

What's hot

Cleaning validation
Cleaning validationCleaning validation
Quality audit plan
Quality audit planQuality audit plan
Quality audit plan
Pravin Jadhao
 
qualification of lcms
qualification of lcmsqualification of lcms
qualification of lcms
Ankush Sule
 
Cleaning validation
Cleaning validationCleaning validation
Cleaning validation
RavichandraNadagouda
 
validation and calibration of HPLC
validation and calibration of HPLCvalidation and calibration of HPLC
validation and calibration of HPLC
Sakshi Garg
 
Hplc method development
Hplc method developmentHplc method development
Hplc method development
Sai Praveen Reddy
 
DEVIATION, OOS &OOT
DEVIATION, OOS &OOT DEVIATION, OOS &OOT
DEVIATION, OOS &OOT
VikasChauhan217
 
Qualification of instrumets
Qualification of instrumetsQualification of instrumets
Qualification of instrumets
ChowdaryPavani
 
Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
Stefan Holt
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
Sai Praveen Reddy
 
Analytical Method Validation.pptx
Analytical Method Validation.pptxAnalytical Method Validation.pptx
Analytical Method Validation.pptx
Bholakant raut
 
Analytical Method Validation basics by Dr. A. Amsavel
Analytical Method Validation  basics by Dr. A. AmsavelAnalytical Method Validation  basics by Dr. A. Amsavel
Analytical Method Validation basics by Dr. A. Amsavel
Dr. Amsavel A
 
Computer system validations
Computer system validationsComputer system validations
Computer system validations
Amruta Balekundri
 
Analytical procedures life cycle management
Analytical procedures life cycle managementAnalytical procedures life cycle management
Analytical procedures life cycle management
Chandra Prakash Singh
 
Analytical methods validation as per ich & usp
Analytical methods validation as per ich & uspAnalytical methods validation as per ich & usp
Analytical methods validation as per ich & usp
GANESH NIGADE
 
Handling deviations & unexpected results during method validation
Handling deviations & unexpected results during method validationHandling deviations & unexpected results during method validation
Handling deviations & unexpected results during method validation
Institute of Validation Technology
 
Qualification of laboratory equipments by Mayuri Soni
Qualification of laboratory equipments by Mayuri SoniQualification of laboratory equipments by Mayuri Soni
Qualification of laboratory equipments by Mayuri Soni
Mayuri Soni
 
Computerized system validation
Computerized system validationComputerized system validation
Computerized system validation
Devipriya Viswambharan
 
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
Kushal Shah
 
Analytical method validation as per ich and usp
Analytical method validation as per ich and usp Analytical method validation as per ich and usp
Analytical method validation as per ich and usp
shreyas B R
 

What's hot (20)

Cleaning validation
Cleaning validationCleaning validation
Cleaning validation
 
Quality audit plan
Quality audit planQuality audit plan
Quality audit plan
 
qualification of lcms
qualification of lcmsqualification of lcms
qualification of lcms
 
Cleaning validation
Cleaning validationCleaning validation
Cleaning validation
 
validation and calibration of HPLC
validation and calibration of HPLCvalidation and calibration of HPLC
validation and calibration of HPLC
 
Hplc method development
Hplc method developmentHplc method development
Hplc method development
 
DEVIATION, OOS &OOT
DEVIATION, OOS &OOT DEVIATION, OOS &OOT
DEVIATION, OOS &OOT
 
Qualification of instrumets
Qualification of instrumetsQualification of instrumets
Qualification of instrumets
 
Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
Analytical Method Validation.pptx
Analytical Method Validation.pptxAnalytical Method Validation.pptx
Analytical Method Validation.pptx
 
Analytical Method Validation basics by Dr. A. Amsavel
Analytical Method Validation  basics by Dr. A. AmsavelAnalytical Method Validation  basics by Dr. A. Amsavel
Analytical Method Validation basics by Dr. A. Amsavel
 
Computer system validations
Computer system validationsComputer system validations
Computer system validations
 
Analytical procedures life cycle management
Analytical procedures life cycle managementAnalytical procedures life cycle management
Analytical procedures life cycle management
 
Analytical methods validation as per ich & usp
Analytical methods validation as per ich & uspAnalytical methods validation as per ich & usp
Analytical methods validation as per ich & usp
 
Handling deviations & unexpected results during method validation
Handling deviations & unexpected results during method validationHandling deviations & unexpected results during method validation
Handling deviations & unexpected results during method validation
 
Qualification of laboratory equipments by Mayuri Soni
Qualification of laboratory equipments by Mayuri SoniQualification of laboratory equipments by Mayuri Soni
Qualification of laboratory equipments by Mayuri Soni
 
Computerized system validation
Computerized system validationComputerized system validation
Computerized system 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
 
Analytical method validation as per ich and usp
Analytical method validation as per ich and usp Analytical method validation as per ich and usp
Analytical method validation as per ich and usp
 

Similar to Analytical method- Content, Development, validation, Transfer & Life Cycle Management

Analytical quality by design
Analytical quality by designAnalytical quality by design
Analytical quality by design
Annalisa Forlenza
 
first lecture-Pharmaceutical Analytical chemistry.ppt
first lecture-Pharmaceutical Analytical chemistry.pptfirst lecture-Pharmaceutical Analytical chemistry.ppt
first lecture-Pharmaceutical Analytical chemistry.ppt
Noorelhuda2
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
akshara01
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
akshara01
 
Life cycle of analytical method
Life cycle of analytical methodLife cycle of analytical method
Life cycle of analytical method
Dr. Nandan Dhir Ph.D.
 
Analytical Method Validations & Detailed Method Validation Parameters
Analytical Method Validations & Detailed Method Validation ParametersAnalytical Method Validations & Detailed Method Validation Parameters
Analytical Method Validations & Detailed Method Validation Parameters
BussaSravanKumar
 
Phase Appropriate Method Validation Aryo Boston-Nitto 2
Phase Appropriate Method Validation Aryo Boston-Nitto 2Phase Appropriate Method Validation Aryo Boston-Nitto 2
Phase Appropriate Method Validation Aryo Boston-Nitto 2Aryo Nikopour
 
HPLC validation.ppt
HPLC validation.pptHPLC validation.ppt
HPLC validation.ppt
Priyanka Yadav
 
Validation of Analytical method.ppt
Validation of Analytical method.pptValidation of Analytical method.ppt
Validation of Analytical method.ppt
Priyanka Yadav
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
SurbhiSharma196
 
JPBA published-a platform aQbD approach for multiple methods development
JPBA published-a platform aQbD approach for multiple methods developmentJPBA published-a platform aQbD approach for multiple methods development
JPBA published-a platform aQbD approach for multiple methods developmentJianmei Kochling
 
Method development
Method developmentMethod development
Method development
Gamal Abdel Hamid
 
QA QC Program for Waste Water Analysis ppt
QA QC Program for Waste Water Analysis pptQA QC Program for Waste Water Analysis ppt
QA QC Program for Waste Water Analysis ppt
KrisnaBagtasos1
 
QbD and PAT Presentation
QbD and PAT PresentationQbD and PAT Presentation
QbD and PAT Presentation
sunp994
 
validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...
Sharath Hns
 
5-Scientific Approach to Validation.pptx
5-Scientific Approach to Validation.pptx5-Scientific Approach to Validation.pptx
5-Scientific Approach to Validation.pptx
AllanThomas30
 
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Merck Life Sciences
 
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
MilliporeSigma
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
roshankhetade2
 
Validation of Analytical Methods.pdf
Validation of Analytical Methods.pdfValidation of Analytical Methods.pdf
Validation of Analytical Methods.pdf
anjaneyulu49
 

Similar to Analytical method- Content, Development, validation, Transfer & Life Cycle Management (20)

Analytical quality by design
Analytical quality by designAnalytical quality by design
Analytical quality by design
 
first lecture-Pharmaceutical Analytical chemistry.ppt
first lecture-Pharmaceutical Analytical chemistry.pptfirst lecture-Pharmaceutical Analytical chemistry.ppt
first lecture-Pharmaceutical Analytical chemistry.ppt
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
 
Life cycle of analytical method
Life cycle of analytical methodLife cycle of analytical method
Life cycle of analytical method
 
Analytical Method Validations & Detailed Method Validation Parameters
Analytical Method Validations & Detailed Method Validation ParametersAnalytical Method Validations & Detailed Method Validation Parameters
Analytical Method Validations & Detailed Method Validation Parameters
 
Phase Appropriate Method Validation Aryo Boston-Nitto 2
Phase Appropriate Method Validation Aryo Boston-Nitto 2Phase Appropriate Method Validation Aryo Boston-Nitto 2
Phase Appropriate Method Validation Aryo Boston-Nitto 2
 
HPLC validation.ppt
HPLC validation.pptHPLC validation.ppt
HPLC validation.ppt
 
Validation of Analytical method.ppt
Validation of Analytical method.pptValidation of Analytical method.ppt
Validation of Analytical method.ppt
 
Ich guidelines
Ich guidelinesIch guidelines
Ich guidelines
 
JPBA published-a platform aQbD approach for multiple methods development
JPBA published-a platform aQbD approach for multiple methods developmentJPBA published-a platform aQbD approach for multiple methods development
JPBA published-a platform aQbD approach for multiple methods development
 
Method development
Method developmentMethod development
Method development
 
QA QC Program for Waste Water Analysis ppt
QA QC Program for Waste Water Analysis pptQA QC Program for Waste Water Analysis ppt
QA QC Program for Waste Water Analysis ppt
 
QbD and PAT Presentation
QbD and PAT PresentationQbD and PAT Presentation
QbD and PAT Presentation
 
validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...validation. chemical and biological evaluation, cleaning validation, personal...
validation. chemical and biological evaluation, cleaning validation, personal...
 
5-Scientific Approach to Validation.pptx
5-Scientific Approach to Validation.pptx5-Scientific Approach to Validation.pptx
5-Scientific Approach to Validation.pptx
 
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
 
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
Webinar: Is Phase-Appropriate Validation the Right Choice for your Cell or Ge...
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
Validation of Analytical Methods.pdf
Validation of Analytical Methods.pdfValidation of Analytical Methods.pdf
Validation of Analytical Methods.pdf
 

Recently uploaded

ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIONDACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
DR SETH JOTHAM
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Savita Shen $i11
 
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
bkling
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
MedicoseAcademics
 
How to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for DoctorsHow to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for Doctors
LanceCatedral
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Saeid Safari
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Dr Jeenal Mistry
 
Ocular injury ppt Upendra pal optometrist upums saifai etawah
Ocular injury  ppt  Upendra pal  optometrist upums saifai etawahOcular injury  ppt  Upendra pal  optometrist upums saifai etawah
Ocular injury ppt Upendra pal optometrist upums saifai etawah
pal078100
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
Levi Shapiro
 
POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
touseefaziz1
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
SumeraAhmad5
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
MedicoseAcademics
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
MedicoseAcademics
 
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptxPharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Dr. Rabia Inam Gandapore
 
The Normal Electrocardiogram - Part I of II
The Normal Electrocardiogram - Part I of IIThe Normal Electrocardiogram - Part I of II
The Normal Electrocardiogram - Part I of II
MedicoseAcademics
 
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptxTriangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Dr. Rabia Inam Gandapore
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
Shweta
 
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Oleg Kshivets
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
DR SETH JOTHAM
 

Recently uploaded (20)

ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIONDACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
ACUTE SCROTUM.....pdf. ACUTE SCROTAL CONDITIOND
 
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model SafeSurat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
Surat @ℂall @Girls ꧁❤8527049040❤꧂@ℂall @Girls Service Vip Top Model Safe
 
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
Report Back from SGO 2024: What’s the Latest in Cervical Cancer?
 
Non-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdfNon-respiratory Functions of the Lungs.pdf
Non-respiratory Functions of the Lungs.pdf
 
How to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for DoctorsHow to Give Better Lectures: Some Tips for Doctors
How to Give Better Lectures: Some Tips for Doctors
 
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists  Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
Ozempic: Preoperative Management of Patients on GLP-1 Receptor Agonists
 
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdfAlcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
Alcohol_Dr. Jeenal Mistry MD Pharmacology.pdf
 
Ocular injury ppt Upendra pal optometrist upums saifai etawah
Ocular injury  ppt  Upendra pal  optometrist upums saifai etawahOcular injury  ppt  Upendra pal  optometrist upums saifai etawah
Ocular injury ppt Upendra pal optometrist upums saifai etawah
 
Cervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptxCervical & Brachial Plexus By Dr. RIG.pptx
Cervical & Brachial Plexus By Dr. RIG.pptx
 
Prix Galien International 2024 Forum Program
Prix Galien International 2024 Forum ProgramPrix Galien International 2024 Forum Program
Prix Galien International 2024 Forum Program
 
POST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its managementPOST OPERATIVE OLIGURIA and its management
POST OPERATIVE OLIGURIA and its management
 
heat stroke and heat exhaustion in children
heat stroke and heat exhaustion in childrenheat stroke and heat exhaustion in children
heat stroke and heat exhaustion in children
 
Physiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of TastePhysiology of Special Chemical Sensation of Taste
Physiology of Special Chemical Sensation of Taste
 
Physiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdfPhysiology of Chemical Sensation of smell.pdf
Physiology of Chemical Sensation of smell.pdf
 
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptxPharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
Pharynx and Clinical Correlations BY Dr.Rabia Inam Gandapore.pptx
 
The Normal Electrocardiogram - Part I of II
The Normal Electrocardiogram - Part I of IIThe Normal Electrocardiogram - Part I of II
The Normal Electrocardiogram - Part I of II
 
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptxTriangles of Neck and Clinical Correlation by Dr. RIG.pptx
Triangles of Neck and Clinical Correlation by Dr. RIG.pptx
 
Evaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animalsEvaluation of antidepressant activity of clitoris ternatea in animals
Evaluation of antidepressant activity of clitoris ternatea in animals
 
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
Lung Cancer: Artificial Intelligence, Synergetics, Complex System Analysis, S...
 
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdfBENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
BENIGN PROSTATIC HYPERPLASIA.BPH. BPHpdf
 

Analytical method- Content, Development, validation, Transfer & Life Cycle Management

  • 1. ANALYTICAL METHOD (DEVELOPMENT, VALIDATION, TRANSFER & LIFE CYCLE MANAGEMENT) Prepared by: MD. MIZANUR RAHMAN MIAJEE 1
  • 2. ANALYTICAL METHOD VALIDATION REGULATORY GUIDANCE: • ICH Q2B- Validation of Analytical Procedures: Methodology • USP <1225> Validation of Compendial Procedures • USP <1226> Verification of Compendial Procedures • USP <1220> The Analytical Procedure Lifecycle – Proposed • USP <621> Chromatography • FDA Guidance- Analytical Procedures and Methods Validation for Drugs and Biologics • CDER Reviewer Guidance- Validation of Chromatographic Methods • Other Journal 2
  • 3. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Principle/Scope: A description of the basic principles of the analytical test/technology (i.e., separation, detection); target analyte(s) and sample(s) type (e.g., drug substance, drug product, impurities) • Apparatus/ Equipment: All required qualified equipment and components (e.g., instrument type, detector, column type, dimensions, and alternative column, filter type). 3
  • 4. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Operating Parameters: Qualified optimal settings and ranges (include allowed adjustments supported by compendial sources or development and/or validation studies) critical to the analysis (e.g., flow rate, components temperatures, run time, detector settings, gradient, head space sampler). A drawing with experimental configuration and integration parameters may be used, as applicable. 4
  • 5. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): Reagents/Standards: The following should be listed where applicable: • Description of reagent or standard • Grade of chemical (e.g., USP/NF, American Chemical Society, High Performance or Pressure Liquid Chromatography, or Gas Chromatography and preservative-free) • Source (e.g., USP reference standard, qualified in-house reference material, WHO International Standard/Reference Material, CBER standard) 5
  • 6. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Purity (for pure chemicals only), State (e.g., dried, undried), and concentration • Potencies (where required by CFR, USP) • Storage conditions • Directions for safe use (as per current Safety Data Sheet) • Validated or documented shelf life 6
  • 7. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): Sample Preparation: Procedures (e.g., extraction method, dilution or concentration, desalting procedures and mixing by sonication, shaking or sonication time) for the preparations for individual sample tests. A single preparation for qualitative and replicate preparations for quantitative tests with appropriate units of concentrations for working solutions (e.g., µg/ml or mg/ml) and information on stability of solutions and storage conditions. 7
  • 8. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Standards Control Solution Preparation: Procedures for the preparation and use of all standard and control solutions with appropriate units of concentration and information on stability of standards and storage conditions, including calibration standards, internal standards, system suitability standards, etc. 8
  • 9. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Procedure: A step-by-step description of the method (e.g., equilibration times, and scan/injection sequence with blanks, placeboes, samples, controls, sensitivity solution (for impurity method) and standards to maintain validity of the system suitability during the span of analysis) and allowable operating ranges and adjustments if applicable. 9
  • 10. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • System Suitability: Confirmatory test(s) procedures and parameters to ensure that the system (equipment, electronics, and analytical operations and controls to be analyzed) will function correctly as an integrated system at the time of use. The system suitability acceptance criteria applied to standards controls and samples, such as peak tailing, precision and resolution acceptance criteria, may be required as applicable. 10
  • 11. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Calculations: The integration method and representative calculation formulas for data analysis (standards, controls, samples) for tests based on label claim and specification (e.g., assay, specified and unspecified impurities and relative response factors). This includes a description of any mathematical transformations or formulas used in data analysis, along with a scientific justification for any correction factors used. 11
  • 12. ANALYTICAL METHOD VALIDATION CONTENT OF IDEAL ANALYTICAL METHOD (AS PER FDA): • Data Reporting: A presentation of numeric data that is consistent with instrumental capabilities and acceptance criteria. The method should indicate what format to use to report results (e.g., percentage label claim, weight/weight, and weight/volume) with the specific number of significant figures needed. The American Society for Testing and Materials (ASTM) E29 standard describes a standard practice for using significant digits in test data to determine conformance with specifications. For chromatographic methods, you should include retention times (RTs) for identification with reference standard comparison basis, relative retention times (RRTs) (known and unknown impurities) acceptable ranges and sample results reporting criteria 12
  • 13. ANALYTICAL METHOD VALIDATION GENERAL POINT MUST BE CONSEDERED FOR ANALYTICAL METHOD (FDA Reviewer Guidance): • The sample and standard should be dissolved in the mobile phase. If that is not possible, then avoid using too high level of the organic solvent as compared to the level in the mobile phase. • The sample and standard concentrations should be close if not the same. • The samples should be bracketed by standards during the analytical procedure 13
  • 14. ANALYTICAL METHOD VALIDATION GENERAL POINT CONSEDER FOR ANALYTICAL METHOD: • Filtration of the samples before injection is occasionally observed. Filtration will remove particulates (centrifugation performs the same function) that may clog columns. Adhesion of the analyte to the filter can also happen. This will be of importance especially for low level impurities. Data to validate this aspect should be submitted by the applicant. 14
  • 16. ANALYTICAL METHOD VALIDATION • An analytical procedure is developed to test a defined characteristic of the drug substance or drug product against established acceptance criteria for that characteristic. • Method development should begin with an initial risk assessment and follow with multivariate experiments. Knowledge gained during these studies on the sources of method variation can help you assess the method performance. ANALYTICAL METHOD DEVELOPMENT 16
  • 17. ANALYTICAL METHOD VALIDATION What is QbD A systematic approach to development that begins with predefined objectives and emphasizes product and process understanding and process control, based on sound science and quality risk management. Benefits of Application of QbD Approach to Analytical Methods • Development of a robust method • Applicable throughout the life cycle of the product Regulatory flexibility − • Movements within “Design Space” are not considered a change in method. ANALYTICAL METHOD DEVELOPMENT 17
  • 19. ANALYTICAL METHOD VALIDATION • What Is An ATP? • Analytical Target Profile is a part of QbD which is a predefined, documented statement of the requirements for the quality of the reportable value, which the analytical procedure must generate in order for the procedure to be considered fit for purpose Using Current Industry Acceptance Criteria. The procedure must be able to accurately quantify [drug] in the [FPP] in the presence of [Impurity/degradation product] with the following requirements for the reportable values: Accuracy = 100.0% ± 3.0% and precision ≤ 2.0% relative standard deviation (RSD). ANALYTICAL METHOD DEVELOPMENT 19
  • 20. ANALYTICAL METHOD VALIDATION • What Is in ATP? In ATP Target profile for the analytical method to be predefined. Some example are given below: 1. Sample sonication time: as per QbD different sonicatione time should be considered to get optimum solubility (ATP) 2. Chromatographic Run Time: A target run time should be selected and as per QbD, different run time to be performed to get optimum one. 3. Sample & standard solution concentration in impurity analysis: Concentration of sample is very important in impurity analysis to cover Reporting threshold limit as per ICH Q2B. A target concentration to be selected before development work. ANALYTICAL METHOD DEVELOPMENT 20
  • 21. ANALYTICAL METHOD VALIDATION Parameters that may be evaluated during method development: During early stages of method development, the robustness of methods should be evaluated because this characteristic can help you decide which method you will submit for approval. Other characteristics that may be considered during method development are: • Specificity • Linearity • Accuracy • Precision • Range • Quantitation limit • Detection limit. • Solution Stability ANALYTICAL METHOD DEVELOPMENT 21
  • 22. ANALYTICAL METHOD VALIDATION ANALYTICAL METHOD DEVELOPMENT Describe the sample Establish Goals Consider sample preparation Choose detector Choose chromatographic mode First Run Optimize separation Predict Problems Validate and Release method 22
  • 23. ANALYTICAL METHOD VALIDATION Collection of literature: Analytical method development starts with literature search where in various pharmacopeias like USP, EP, BP, JP etc and chromatographic journals are referred to check the availability of suitable analytical methods. If any suitable method is found, it is still necessary to perform method optimization and validation to prove that the method can be successfully adapted for its intended use. 23
  • 24. ANALYTICAL METHOD VALIDATION Sample information: The synthetic route from raw material to finished dosage forms (structures) of the molecule should be collected and the impurities originating from starting materials of synthesis, degradation products, excipients, solvents etc. need to be considered. The method should be designed based on the closely related structures and get the best resolution between the closely related compounds. The structures of impurities, starting material, intermediates and degradation products are compared with the structure of drug substances and arrive at the polarity whether they are less polar or more polar than the compound of interest. 24
  • 25. ANALYTICAL METHOD VALIDATION • Selection of diluents: The diluent should be chosen based on the solubility of impurities, degradation products, starting materials, intermediates and the analyte. The diluent should be compatible with the mobile phase to get a better peak shape of analyte. 25
  • 26. ANALYTICAL METHOD VALIDATION • Selection of stationary phase: The selection of bonded phase can be based on the polarity of molecule and its by-products. For RP-LC, a wide variety of columns are accessible covering a large range of polarity by cross-linking Si-OH groups with linear alkyl chains like C8, C18 and phenyl groups (-C6H6), nitrile groups (-CN), , different embedded hybridized groups amino groups (-NH2) etc. C18 columns are the commonly used columns in HPLC method analysis. C8 or Octyl bonded phases are also used occasionally. Like C18, they are non-polar, but not as hydrophobic. 26
  • 27. ANALYTICAL METHOD VALIDATION • Detectors: Various detectors used in HPLC instrument include UV-Visible detector, photodiode array detector, fluorescence detector, conductivity detector, refractive index detector, electro- chemical detector, mass spectrometer detector and evaporative light scattering detector. UV-Visible detectors are typical in many laboratories as they can detect a wide array of compounds 27
  • 28. ANALYTICAL METHOD VALIDATION • pH range: Method development within the different pH ranges from 1 to 12 for better chromatographic resolution between two or more peaks of an analyte depends upon three main factors, column efficiency, selectivity and retention time. The ionizable analytes are either bases or acids and they affect the above three factors dramatically with change in pH. Retention time can be improved by changing the pH that will lead to easy separation of ionizable analytes from non-ionized forms. 28
  • 29. ANALYTICAL METHOD VALIDATION • pH range: To achieve optimum resolution, it requires change in the pH of mobile phase. Method development can proceed by investigating parameters of chromatographic separations first at low pH and then at higher pH until optimum results are achievd. 29
  • 30. ANALYTICAL METHOD VALIDATION • Mobile phase composition: Mobile phase composition (or solvent strength) plays an important role in RP-HPLC separation. Acetonitrile (ACN), methanol (MeOH) and tetrahydrofuran (THF) are commonly used solvents in RP -HPLC having low UV cut- off of 190, 205 and 212nm respectively. These solvents are miscible with water. Mixture of acetonitrile and water is the best initial choice for the mobile phase during method development. 30
  • 31. ANALYTICAL METHOD VALIDATION • Column temperature: Separation of many samples can be enhanced by selecting the right column temperature. Higher column temperature reduces system backpressure by decreasing mobile phase viscosity, which in turn allows use of longer columns with higher separation efficiency. The optimum temperature is dependent upon the nature of the mixture components. The overall separation can be improved by the simultaneous changes in column temperature and mobile phase composition 31
  • 32. ANALYTICAL METHOD VALIDATION • Analytical Method Transfer: What is analytical method transfer: It is the documented process that qualifies a laboratory (the receiving unit) to use an analytical test procedure that originated in another laboratory (the transferring unit), thus ensuring that the receiving unit has the procedural knowledge and ability to perform the transferred analytical procedure as intended. 32
  • 33. ANALYTICAL METHOD VALIDATION • Transfer may be- • One R & D development lab to another R & D development lab • R & D development lab to quality control lab inside same unit • R & D development lab to quality control lab of other unit • One facility to another intra-company facility • One company to another company such as a contract research organization 33
  • 34. ANALYTICAL METHOD VALIDATION Procedure of Analytical Method Transfer: Analytical Method Transfer (AMT) can be done by two ways: 1. Comparative Testing 2. Co-validation between Two or More Laboratories 34
  • 35. ANALYTICAL METHOD VALIDATION 1. Comparative Testing: analysis of a predetermined number of samples of the same lot by both the sending and the receiving units. Prerequisite: A preapproved transfer protocol prepared by sending unit that stipulates the details of the procedure, the samples that will be used, and the predetermined acceptance criteria, including acceptable variability Criteria: Meeting the predetermined acceptance criteria is necessary to assure that the receiving unit is qualified to run the procedure. 35
  • 36. ANALYTICAL METHOD VALIDATION 2. Co-validation Between Two or More Laboratories: The transferring unit can involve the receiving unit in an inter laboratory co-validation, including them as a part of the validation team at the transferring unit and thereby obtaining data for the assessment of reproducibility Prerequisite: a preapproved transfer or validation protocol that provides the details of the procedure, the samples to be used, and the predetermined acceptance criteria Criteria: Meeting the predetermined acceptance criteria is necessary to assure that the receiving unit is qualified to run the procedure. 36
  • 37. ANALYTICAL METHOD VALIDATION Transfer Waiver: • The new product’s composition is comparable to that of an existing product and/or the concentration of active ingredient is similar to that of an existing product and is analyzed by procedures with which the receiving unit already has experience • The analytical procedure being transferred is described in the USP–NF, and is unchanged. Verification should apply in this case • The analytical procedure transferred is the same as or very similar to a procedure already in use. 37
  • 38. ANALYTICAL METHOD VALIDATION Transfer Waiver: • The personnel in charge of the development, validation, or routine analysis of the product at the transferring unit are moved to the receiving unit. If eligible for transfer waiver, the receiving unit should document it with appropriate justifications. 38
  • 39. ANALYTICAL METHOD VALIDATION • ELEMENTS RECOMMENDED FOR THE TRANSFER OF ANAYTICAL PROCEDURES: • Transferring unit: • provide training to the receiving unit, training should be documented, providing the analytical procedure, the reference standards, the validation reports, and any necessary documents, as well as for providing the necessary training and assistance to the receiving unit as needed during the transfer. 39
  • 40. ANALYTICAL METHOD VALIDATION • ELEMENTS RECOMMENDED FOR THE TRANSFER OF ANAYTICAL PROCEDURES: • Receiving unit: • Provides qualified staff or properly trains the staff before the transfer, ensures that the facilities and instrumentation are properly calibrated and qualified as needed, and verifies that the laboratory systems are in compliance with applicable regulations and in-house general laboratory procedures. 40
  • 41. ANALYTICAL METHOD VALIDATION • ELEMENTS RECOMMENDED FOR THE TRANSFER OF ANAYTICAL PROCEDURES: • Both unit: Compare and discuss data as well as any deviations from the protocol. This discussion addresses any necessary corrections or updates to the final report and the analytical procedure as necessary to reproduce the procedure. A single lot of the article 41
  • 42. ANALYTICAL METHOD VALIDATION • ELEMENTS RECOMMENDED FOR THE TRANSFER OF ANAYTICAL PROCEDURES: • Content of transfer protocol: Below mentioned parameters must be included in transfer protocol • Objective • Scope • Responsibilities of the transferring and receiving units, • Materials and instruments that will be used, • Analytical procedure • Experimental design • Acceptance criteria for all tests and/or methods included in the transfer. 42
  • 43. ANALYTICAL METHOD VALIDATION • Analytical Procedure for AMT: The procedure should be written with sufficient detail and explicit instructions, so that a trained analyst can perform it without difficulty. A pre- transfer meeting between the transferring and receiving units is helpful to clarify any issues and answer any questions regarding the transfer process. If complete or partial validation data exist, they should be available to the receiving unit, along with any technical details required to perform the test in question. 43
  • 44. ANALYTICAL METHOD VALIDATION • What is analytical method validation Validation is the process of demonstrating that an analytical procedure is suitable for its intended purpose 44
  • 45. ANALYTICAL METHOD VALIDATION • Importance of Analytical Method Validation  Validation of a method involves using experimental design to prove that the method can produce accurate and precise results within the scope of its intended use  Understanding the application and limitations of the test method will allow for accurate assessment of sample information, ranging from process outputs to commercial release testing and many steps in between.  If the method validation has not been performed or has been performed in an inadequate manner, the method is not proven to provide reliable data. • A majority of audit findings fell into three main categories: 1. the use of a non- validated method for critical decision making; 2. inadequate method validation that did not provide the necessary information; 3. method validation that lacked appropriate controls to maintain the integrity of the validation 45
  • 46. ANALYTICAL METHOD VALIDATION • Types of Analytical Method need to validate  Identification tests  Quantitative tests for impurities;  Limit tests for the control of impurities; and  Assay tests for the active component in a drug substance, drug product, or other selected component(s) in the drug product. 46
  • 47. ANALYTICAL METHOD VALIDATION • Noncompendial Analytical Procedures: Requires Complete Validation • Compendial Analytical Procedures: Requires Verification 47
  • 48. ANALYTICAL METHOD VALIDATION • Why verification:  Drug substance's synthetic route,  The method of manufacture for the drug product  Drug substances from different suppliers may have different impurity profiles that are not addressed by the compendial test procedure  The excipients in a drug product can vary widely among manufacturers and may have the potential to directly interfere with the procedure or cause the formation of impurities that are not addressed by the compendia procedure  Drug products containing different excipients, antioxidants, buffers, or container extractives may affect the recovery of the drug substance from the matrix (But as per current thinking of FDA, FDA prefer complete validation for compendia method instead of verification) 48
  • 49. ANALYTICAL METHOD VALIDATION • Following parameter to be considered for Analytical Method Validation:  Specificity  Accuracy  Precision (Repeatability, Intermediate Precision & Reproducibility)  Linearity  Range  Quantitation limit (LOQ)  Detection limit (LOD)  Robustness 49
  • 50. ANALYTICAL METHOD VALIDATION SPECIFICITY /SELECTIVITY Ability to assess unequivocally the analyte in the presence of components that may be expected to be present, such as impurities, degradation products, and matrix components. • Identification: Ensure the identity of the analyte. • Impurity: Ensure that all of the analytical procedures performed allow an accurate statement of the content of impurities of an analyte (e.g., related substances test, heavy metals limit, or organic volatile impurities). • Assays: Provide an exact result, which allows an accurate statement on the content or potency of the analyte in a sample 50
  • 51. ANALYTICAL METHOD VALIDATION SPECIFICITY /SELECTIVITY • Procedure: When Impurities are available (Specified identified impurity): 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 (by comparison with the assay result obtained on unspiked samples). For the impurity test, the discrimination may be established by spiking drug substance or drug product with appropriate levels of impurities and demonstrating the separation of these impurities individually and/or from other components in the sample matrix. 51
  • 52. ANALYTICAL METHOD VALIDATION Accuracy The accuracy of an analytical procedure is the closeness of test results obtained by that procedure to the true value. Evaluate by analyzing synthetic mixtures: known quantity of API with excipients/ Impurity • Recommended Data: Accuracy should be assessed using a minimum of 9 determinations over a minimum 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 percent 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. 52
  • 53. ANALYTICAL METHOD VALIDATION Precision The precision of an analytical procedure is the degree of agreement among individual test results when the procedure is applied repeatedly to multiple samplings of a homogeneous sample. The precision of an analytical procedure is usually expressed as the standard deviation or relative standard deviation (coefficient of variation) of a series of measurements A. Repeatability • Repeatability should be assessed using: (1) A minimum of 9 determinations covering the specified range for the procedure (e.g., 3 concentrations/3 replicates each); or (2) A minimum of 6 determinations at 100% of the test concentration. 53
  • 54. ANALYTICAL METHOD VALIDATION Precision B. Intermediate Precision 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 necessary to study these effects individually. The use of an experimental design (matrix) is encouraged. 54
  • 55. ANALYTICAL METHOD VALIDATION Precision C. 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 55
  • 56. ANALYTICAL METHOD VALIDATION Linearity & Range Linearity: The linearity of an analytical procedure is its ability to elicit test results that are directly, or by a well-defined mathematical transformation, proportional to the concentration of analyte in samples within a given range Parameter to check: The correlation coefficient, y-intercept, slope of the regression line, and residual sum of squares should be submitted. Number of sample/ Injection: For the establishment of linearity, a minimum of five concentrations is recommended. Other approaches should be justified. 56
  • 57. ANALYTICAL METHOD VALIDATION Linearity & Range Range: The range of an analytical procedure is the interval between the upper and lower levels of analyte (including these levels) that have been demonstrated to be determined with a suitable level of precision, accuracy, and linearity using the procedure as written assay of a drug substance or a finished (drug) product: Normally from 80% to 120% of the test concentration For content uniformity: Covering a minimum of 70% to 130% of the test concentration, unless a wider, more appropriate range, based on the nature of the dosage form (e.g., metered dose inhalers), is justified 57
  • 58. ANALYTICAL METHOD VALIDATION Linearity & Range • 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 • For the determination of an impurity: From the reporting level of an impurity to 120% of the specification. For impurities known to be unusually potent or to produce toxic or unexpected pharmacological effects, the detection/quantitation limit should be commensurate with the level at which the impurities must be controlled. • If assay and purity are performed together as one test and only a 100% standard is used, linearity should cover the range from the reporting level of the impurities to 120% of the assay specification. 58
  • 59. ANALYTICAL METHOD VALIDATION Detection limit It is the lowest amount of analyte in a sample that can be detected, but not necessarily quantitated, under the stated experimental conditions. Thus, limit tests merely substantiate that the amount of analyte is above or below a certain level. The detection limit is usually expressed as the concentration of analyte (e.g., percentage or parts per billion) in the sample. A. 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 59
  • 60. ANALYTICAL METHOD VALIDATION Detection limit B. 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. Signal-to-noise ratio between 3 or 2:1 is generally considered acceptable for estimating the detection limit. 60
  • 61. ANALYTICAL METHOD VALIDATION Detection limit C. Based on the Standard Deviation of the Response and the Slope The detection limit (DL) may be expressed as: where = 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. The estimate of may be carried out in a variety of ways, for example: 61
  • 62. ANALYTICAL METHOD VALIDATION Detection limit 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. 62
  • 63. ANALYTICAL METHOD VALIDATION Detection limit 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. 63
  • 64. ANALYTICAL METHOD VALIDATION Quantitation Limit The quantitation limit is a characteristic of quantitative assays for low levels of compounds in sample matrices, such as impurities in bulk drug substances and degradation products in finished pharmaceuticals. It is the lowest amount of analyte in a sample that can be determined with acceptable. Precision and Accuracy under the stated experimental conditions. The quantitation limit is expressed as the concentration of analyte (e.g., percentage or parts per billion) in the sample. QL to be determined as like Detection limit except signal-to-noise ratio is 10:1. and the calculation would be 64
  • 65. ANALYTICAL METHOD VALIDATION Quantitation Limit 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 65
  • 66. ANALYTICAL METHOD VALIDATION Robustness The evaluation of robustness should be considered during the development phase and depends on the type of procedure under study. It should show the reliability of an analysis with respect to deliberate variations in method parameters. 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. 66
  • 67. ANALYTICAL METHOD VALIDATION Robustness • Examples of typical variations are: o Stability of analytical solutions o Extraction time • In the case of liquid chromatography, examples of typical variations are: o Influence of variations of pH in a mobile phase o Influence of variations in mobile phase composition o Different columns (different lots and/or suppliers) o Temperature o Flow rate 67
  • 68. ANALYTICAL METHOD VALIDATION Robustness System suitability testing: These tests are used to verify that the chromatographic system is adequate for the intended analysis. system suitability tests are performed by collecting data from replicate injections of standard or other solutions as specified in the individual monograph. • Factors that may affect chromatographic behavior include the following: Composition, ionic strength, temperature, and apparent pH of the mobile phase• o Flow rate, column dimensions, column temperature, and pressure o Stationary phase characteristics, including type of chromatographic support (particle-based or monolithic), particle or macropore size, porosity, and specific surface area o Reverse-phase and other surface modification of the stationary phases, the extent of chemical modification (as expressed by end-capping, carbon loading, and others) 68
  • 69. ANALYTICAL METHOD VALIDATION Robustness System Suitability: Unless otherwise specified in the individual monograph, data from five replicate injections of the analyte are used to calculate the relative standard deviation (RSD), if the requirement is ≤ 2.0%; data from six replicate injections are used if the RSD requirement is >2.0%. If adjustments of operating conditions are necessary in order to meet system suitability requirements, each of the items in the following list is the maximum variation that can be considered: 69
  • 70. ANALYTICAL METHOD VALIDATION Robustness pH of mobile phase (HPLC): ±0.2 units of the value or range specified. Concentration of salts in buffer (HPLC): ±10% if the permitted pH variation Ratio of components in mobile phase (HPLC): ±30% relative. However, the change in any component cannot exceed ±10% absolute (i.e., in relation to the total mobile phase) Wavelength of UV-visible detector (HPLC): ±3 nm Stationary phase: Length- between −25% and 50% of the prescribed L/dp Flow rate (HPLC): ±50% (Based on other factors) Injection volume (HPLC): Can be adjusted as far as it is consistent with accepted precision, linearity, and detection limits Column temperature (HPLC): As much as ±10°. 70
  • 71. ANALYTICAL METHOD VALIDATION Life Cycle Management of Analytical Procedure Once an analytical procedure (including compendial methods) is successfully validated or verified and implemented, the procedure should be followed during the life cycle of the product to continually assure that it remains fit for its intended purpose. 71
  • 72. ANALYTICAL METHOD VALIDATION Life Cycle Management of Analytical Procedure What to be done:  Trend analysis on method performance at regular intervals  New information and risk assessments (e.g., a better understanding of product CQAs or awareness of a new impurity) may warrant the development and validation of a new or alternative analytical method  An appropriate number of retention samples should be maintained to allow for comparative studies.  The retention samples used in comparative studies should include samples that represent marketed product and, when possible, pivotal clinical trial material. 72
  • 73. ANALYTICAL METHOD VALIDATION Life Cycle Management of Analytical Procedure Re-Validation:  When a change is made to an analytical procedure, such as a change in equipment or reagent, then re-validation of all or part of the analytical procedure needs to be considered.  Method re-validation should focus on the critical performance characteristics of the method, such as specificity, precision, and accuracy.  The scope of re-validation should be risk-based. 73
  • 74. ANALYTICAL METHOD VALIDATION Life Cycle Management of Analytical Procedure • Analytical Method Comparability Studies: Alternative Analytical Procedures An alternative analytical procedure is an analytical procedure that is used in place of the FDA approved analytical procedure. After approval, for an NDA or ANDA, or for a procedure approved in a BLA but not included in an FDA regulation, the addition, revision, or deletion of an alternative analytical procedure that provides the same or increased assurance of the identity, strength, quality, purity, or potency of the material being tested as the analytical procedure described in the approved application, must be documented in the next annual report 74
  • 75. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) If a procedure is a validated quantitative analytical procedure that can detect changes in a quality attribute(s) of the drug substance and drug product during storage, it is considered a stability- indicating test. To demonstrate specificity of a stability-indicating test, a combination of challenges should be performed. Some challenges include the use of samples spiked with target analytes and all known interferences; samples that have undergone various laboratory stress conditions; and actual product samples (produced by the final manufacturing process) that are either aged or have been stored under accelerated temperature and humidity conditions. 75
  • 76. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) Objective The main objective of a stability indicating method is to monitor results during stability studies in order to guarantee safety, efficacy and quality. It represents also a powerful tool when investigating out-of-trend (OOT) or out-of-specification (OOS) results in quality control processes. Important factor to consider: The method should be sensitive to the reportable impurity level, LOQ (Limit of Quantitation), which is typically 0.05% of label claim, should be established in the method, and the method should be linear from LOQ to typically up to 150% of the nominal standard (STD) concentration 76
  • 77. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) 77
  • 78. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) • How develop a SIM: Generation of degraded samples for testing selectivity of the method Determination of Limit of Quantification (LoQ) In close relation to the determination of the amount of degradation is the evaluation of Limit of Detection (LoD) and Limit of Quantification (LoQ) of the method. These limits should be closely related to the Reporting, Identification and Qualification of degradation products, as stated in ICH Q3B (R2). These thresholds are determined either as percentage of drug substance or total daily intake (TDI) of degradation product. 78
  • 79. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) • Force Degradation: Objective of forced degradation studies:  To establish degradation pathways of drug substances and drug products.  To differentiate degradation products that are related to drug products from those that are generated from non-drug product in a formulation.  To elucidate the structure of degradation products.  To determine the intrinsic stability of a drug substance in formulation.  To reveal the degradation mechanisms such as hydrolysis, oxidation, thermolysis or photolysis of the drug substance and drug product  To establish stability indicating nature of a developed method.  To understand the chemical properties of drug molecules.  To generate more stable formulations.  To produce a degradation profile similar to that of what would be observed in a formal stability study under ICH conditions. • To solve stability-related problems 79
  • 80. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) 80
  • 81. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) What to include: The testing should include the effect of temperatures (in 10°C increments (ie, 50°C, 60°C) above that for accelerated testing), humidity (ie, 75% relative humidity or greater) where appropriate, oxidation, and photolysis on the drug substance. The testing should also evaluate the susceptibility of the drug substance to hydrolysis across a wide range of pH values when in solution or suspension.” 81
  • 82. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) 82
  • 83. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) How much is enough Values anywhere between 5% to 20% degradation of the drug substance have been considered as reasonable and acceptable for validation of chromatographic assays. However, for small pharmaceutical molecules for which acceptable stability limits of 90% of label claim is common, pharmaceutical scientists have agreed that approximately 10% degradation is optimal for use in analytical validation. 83
  • 84. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) Selection of drug concentration Which concentration of drug should be used for degradation study has not been specified in regulatory guidance. It is recommended that the studies should be initiated at a concentration of 1 mg/mL . By using drug concentration of 1 mg/mL, it is usually possible to get even minor decomposition products in the range of detection 84
  • 85. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) What is peak purity: Peak purity is an analysis of spectral differences, assuming the impurities are spectrally different from the analyte. The spectral analysis is achieved using vector analysis algorithms The more similar the spectra are, the closer the value is to 0.0 degrees; the more spectrally different, the larger the value (90 degrees is the maximum). When no significant spectral differences are found within a peak, one can conclude that the peak is spectrally homogeneous and probably contains a single compound. 85
  • 86. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM)  Peak Purity algorithm analyzes all spectra within a peak  The Apex Spectrum is the reference spectrum that it compares to.  If Purity Angle < Purity Threshold, Within the noise of the system the peak is spectrally homogeneous or spectrally pure.  If Purity Angle > Purity Threshold , there is something within the peak that cannot be explained by noise. The peak is not spectrally pure, the Purity Flag field is checked. 86
  • 87. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) Purity Angle > Purity Threshold = Impure Peak 87
  • 88. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) Purity threshold > Purity Angle = Pure Peak 88
  • 89. ANALYTICAL METHOD VALIDATION Stability Indicating Method (SIM) Mass balance • Mass balance correlates the measured loss of a parent drug to the measured increase in the amount of degradation products. It is a good quality control check on analytical methods to show that all degradation products are adequately detected and do not interfere with quantitation of the parent drug (i.e.,stability-indicating methods). 89
  • 90. ANALYTICAL METHOD VALIDATION System Suitability General Recommendation: System suitability testing is essential for the assurance of the quality performance of the chromatographic system. The amount of testing required will depend on the purpose of the test method. For dissolution or release profile test methods using an external standard method, k', T and RSD are minimum recommended system suitability tests. For acceptance, release, stability, or impurities / degradation methods using external or internal standards, k', T, R, and RSD are recommended as minimum system suitability testing parameters. 90
  • 91. ANALYTICAL METHOD VALIDATION System Suitability Capacity factor (k'): k' = (tR - to) /to The capacity factor is a measure of where the peak of interest is located with respect to the void volume, i.e., elution time of the non-retained components. Recommendations: The peak should be well-resolved from other peaks and the void volume. Generally the value of k' is > 2. Void volume: amount of dead volume in the column that is not taken up by the particle size of the stationary phase 91
  • 92. ANALYTICAL METHOD VALIDATION System Suitability Resolution (R,): R, is a measure of how well two peaks are separated. For reliable quantitation, well-separated peaks are essential for quantitation. This is a very useful parameter if potential interference peak(s) may be of concern. The closest potential eluting peak to the R, is minimally influenced by the ratio of the two compounds being measured. The resolution of peaks as indicated by the R, values is shown in below Figure: Recommendations: R, of > 2 between the peak of interest and the closest potential interfering peak (impurity, excipient, degradation product, internal standard, etc.) is desirable. 92
  • 93. ANALYTICAL METHOD VALIDATION System Suitability Tailing factor (T): The accuracy of quantitation decreases with increase in peak tailing because of the difficulties encountered by3the integrator in determining wheretwhen the peal< ends and hence the calculation of the area under the peal peak of interest. Below Figures illustrate the tailing factors and the effect on quantitation. If the integrator is unable to determine exactly when an upslope or downslope occurs, ' accuracy drops. Recommendations: T of less than 2 93
  • 94. ANALYTICAL METHOD VALIDATION System Suitability Theoretical plate number (N): Theoretical plate number is a measure of column efficiency, that is, how many peaks can be located per unit run-time of the chromatogram. N is fairly constant for each peak on a chromatogram with a fixed set of operating conditions. Recommendations: The theoretical plate number depends on elution time but in general. should be > 2000. 94
  • 95. ANALYTICAL METHOD VALIDATION System Suitability Precision /injection repeatability (RSD): Injection precision expressed as RSD (relative standard deviation) indicates the performance of the HPL chromatograph which includes the plumbing, column, and environmental conditions, the time the samples are analyzed. It should be noted that sample preparation and manufacturing variations are not considered. Recommendations: RSD of s 1% is desirable. 95
  • 96. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 96
  • 97. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 97
  • 98. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 98
  • 99. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 99
  • 100. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 100
  • 101. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 101
  • 102. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 102
  • 103. ANALYTICAL METHOD VALIDATION Example of FDA Warning letter 103