SlideShare a Scribd company logo
Phase Appropriate Method Validation
Aryo Nikopour
The Southern California Pharmaceutical Discussion Group (SCPDG) of AAPS
Irvine, CA
January 12, 2017
VALIDATION OF ANALYTICAL METHOD
DATA QUALITY TRIANGLE
METHOD LIFE CYCLE
Validation
Development Optimization
METHOD VALIDATION
PUBLISHED VALIDATION GUIDELINES
 1978 Current Good Manufacturing Practices (cGMP)
 1987 FDA Validation Guideline
 1989 Supplement 9 to USP XXI
 1994 CDER Reviewer Guidance: Validation of Chromatographic Method
 1995 ICH Validation Definitions: Q2A, Text on Validation of Analytical procedures
 1997 ICH Validation Methodology: Q2B, Validation of Analytical Procedures: Methodology
 1999 Supplement 10 to USP 23 <1225>: Validation of Compendial Methods
 1999 CDER “Bioanalytical Method Validation for Human Studies”
 2000 CDER Draft “Analytical Procedures and Method Validation”
 2014 CDER/CBER Guidance for Industry: “Analytical Procedure and Method Validation for Drug
and Biologic”
 PDA Technical Report No. 57 : Analytical Method Validation and Transfer for Biotechnology
Products”
GUIDELINES FOR METHOD VALIDATION
www.ICH.orgwww.ICH.org
(Dates indicate ICH finalization)
ICH Q2(R1): Validation of Analytical Procedures:
Methodology- Nov. 2005
ICH Q3A(R): Impurities in New Drug Substances - Feb. 2002
ICH Q3B(R): Impurities in New Drug Products – Feb. 2003
ICH Q3C: Impurities: Residual Solvents - July 1997
ICH Q5A,D: Biotech/Biological Products - 1997
ICH Q5B,C: Biotech/Biological Products - 1995
GUIDELINES FOR METHOD VALIDATION
www.ICH.orgwww.ICH.org
(Dates indicate ICH finalization)
ICH Q2(R1): Validation of Analytical Procedures:
Methodology- Nov. 2005
ICH Q3A(R): Impurities in New Drug Substances - Feb. 2002
ICH Q3B(R): Impurities in New Drug Products – Feb. 2003
ICH Q3C: Impurities: Residual Solvents - July 1997
ICH Q5A,D: Biotech/Biological Products - 1997
ICH Q5B,C: Biotech/Biological Products - 1995
VALIDATION IN THE 21 CENTURY
METHOD VALIDATION
CFR
• There are many reason to validate analytical methods:
– Regulatory Requirements
– Good Science
– Quality Control requirements.
• The Code of Federal Regulations (CFR) 311.165c explicitly states that the,
“Accuracy, Sensitivity, Specificity, and Reproducibility of test methods
employed by the firm shall be established and documented.”
ICH GUIDELINE Q2(R1)
• The objective of validation of an analytical procedure is to
demonstrate that it is suitable for its intended purpose,
In practice, it is usually possible to design the experimental
work such that the appropriate validation characteristics can
be considered simultaneously, to provide a sound, overall
knowledge of the capabilities of the analytical procedure, for
instance; Specificity, Linearity, Range, Accuracy, and
Precision.
Support the identity, strength, quality, purity, and potency
of the drug substances and drug products.
WHAT IS METHOD VALIDATION?
 Validation is procedure dependent.
 Validation, “Proves” the procedure works as described.
 Validation is product specific.
 Procedures are instrument dependent.
VERIFICATION USP <1226>
• Current USP <1226> Verification of Compendial Procedure
– The Analytical procedures in the current USP are legally recognized under section
501(b) of the Federal Food, Drug and Cosmetic Act as the regulatory analytical
procedures for the compendial items. The suitability of these procedures must be
verified under the actual conditions of use.
VERIFICATION
• When using USP analytical procedures, the guidance recommends
that information be provided for the following characteristics:
– Specificity of the procedure
– Stability of the sample solution
– Intermediate precision
METHOD TRANSFER, USP <1224>
• Method Transfer is a documented process that qualifies a
laboratory (Receiving Lab) to use an analytical test
procedure that is originated from the transferring laboratory.
• Types of Method Transfer:
– Comparative Testing
– Co -validation
– Revalidation/Partial Validation
– Transfer Waiver
CLASSIFICATION OF VALIDATED
ANALYTICAL METHODS
Compendial (USP 39/ NF 34):
• Legally recognized under section 501 (b) of the Federal Food, Drug, and Cosmetic Act.
• Recommends information be provided for; Specificity, Sample Solution Stability, and
Intermediate Precision.
Noncompendial:
• Submitted with the NDA/ BLA or ANDA application.
• If the compendial procedure is not stability-indicating, perform an alternative analytical
procedure with complete validation.
USP <1225>ASSAY CATEGORIES
CategoryCategory
NumberNumber Category NameCategory Name Description of AssayDescription of Assay
I Quantitative
Quantitation of major
components/active ingredients
present at high concentrations.
II
Impurities-
Quantitative Determination of impurities or
degradation products.
II Impurities-Limit
III
Performance
Characteristics
Parameters to be tested depend on
the nature of the test; includes
dissolution testing.
IV Identity
METHOD VALIDATION REQUIREMENTS
USP Assay Category
I
II
III IV
Parameter: Quantitative Limit Tests
Accuracy Y Y • Y N
Precision Y Y N Y N
Intermediate
Precision
Y Y N Y N
Specificity N Y Y N Y
Detection Limit N Y Y • N
Quantitation Limit N Y N • N
Linearity Y Y N • N
Range Y Y • • N
Robustness Y Y N N N
Selectivity Y Y N Y Y
System Suitability Y Y N Y N
Solution Stability Y Y N Y N
• May be required, depending on the nature of the specific test.
PHASE APPROPRIATE VALIDATION
Pre-
clinical PM
METHOD VALIDATION READINESS
Define the application, purpose and scope of the method.
Define Analytes, Dosage Strength and Sample Matrix.
Review Method Development Summary Report.
Evaluate method validation parameters during development.
METHOD VALIDATION CHARACTERISTICS
Validation Characteristics Experimental Details Acceptance Criteria
Specificity Stress Studies 5-10% Degradation
Selectivity Determine Chromatographic non-
interference
No inference , minimum resolution
between peaks of interest and impurities
should be >1.5
System Suitability System precision assessed by 6 replicate
measurement/injections
%RSD ≤2%
Linearity At least 5 Concentration over the range
Assay: 50% to 125% of Specification limit
QL-150% of specification limit
Calibration Model is valid
R ≥0.998
Report Intercept, Slope and %Bias
Detection Limit (DL) DL= 3.3 (DL= 3.3 (σσ/S)/S) S/N≥ 3S/N≥ 3
Quantitation Limit (QL) DL= 10 (DL= 10 (σσ/S)/S) %RSD≤ 15%%RSD≤ 15%
METHOD VALIDATION CHARACTERISTICS
Validation Characteristics Experimental Details Acceptance Criteria
Precision :
Repeatability
Intermediate Precision (Ruggedness)
Reproducibility
6 replicates
6 replicates
%RSD≤ 2%
Overall %RSD (two Analyst)
Accuracy At least 9 determination over 3
concentration level
e.g. 70 to 120% for
For Assay Mean Recovery 97 to 103%
for Impurities : 85% to 115%
Range The range is defined by the results
obtained for linearity, accuracy and
precision
Linearity, accuracy and precision
demonstrated over the range
Solution Stability Determine solution stability of Reference
Standard Solution and Sample over 72
hours
98 to 102 % of control
Robustness Deliberately change critical parameters of
the method
Must meet system suitability and
selectivity requirements
VALIDATION: PHASE I
Drug Product Assay I.D.
Quantitative
Impurities
Limit Test
Selectivity X X X X
Repeatability X X
Accuracy/Precision Recovery at
100%
At 100% of
Reporting
Threshold
Linearity X QL to 200% of
Limit
Range Defined by ALP Defined by ALP
DL/QL DL QL QL or at Limit
System Suitability X X X X
Solution Stability X X X
VALIDATION: PHASE II
Assay I.D Quantitative
Impurities
Limit Test
Selectivity X X X X
Specificity X
Repeatability X X X
Accuracy Recovery at 3
levels
At 100% of
Reporting
Threshold
Linearity X X X
DL/QL DL X QL
Range Define by ALP Defined by ALP
System Suitability X X X X
Solution Stability X X X
VALIDATION: PHASE III
Assay I.D Quantitative
Impurities
Limit Test
Selectivity X X X X
Specificity X
Repeatability X X X
Intermediate Precision X 2nd
Analyst X X
Accuracy X X
Linearity X X
DL/QL DL X QL
Range Defined by ALP Defined by ALP
Solution Stability X X X
System Suitability X X X X
Robustness X X X
METHOD VALIDATION
SYSTEM SUITABILITY
Based on the concept that the equipment, electronics, analytical operations and
samples to be analyzed constitute an integral system that can be evaluated as such.
What parameters do you measure forWhat parameters do you measure for
system suitabilitysystem suitability
SYSTEM SUITABILITY
What parameters do you measure for system suitability?What parameters do you measure for system suitability?
CapacityCapacityEfficiencyEfficiencySelectivitySelectivity
Date NB/Page
Standard B1 (n=6) Injections SST Solution B1 and B2
Area %RSD
Average Tailing
Factor %RSD Retention Time Average Theoretical Plates
Resolution LVF and
DesMethyl-LVF)
Response Factor %
Difference
≤ 1% 0.8 ≤ Tf ≤ 1.4 ≤ 1% >15000 NLT 2.5 ≤ 2%
8/18/2006 1494/18 0.1 1.03 0.1 29834 3.27 0.2
8/18/2006 1494/31 0.1 1.03 0 32177 3.28 0.1
8/20/2006 1494/52 0.1 1.03 0.1 27792 3.3 0.7
8/22/2006 1494/72 0.1 1.03 0 26567 3.31 2.7
8/23/2006 1504/1 0 1.03 0.2 27228 3.29 0.8
8/24/2006 1504/8 0.2 1.02 0.1 26535 3.32 1.2
8/25/2006 1504/17 0.1 1.02 0.1 26903 3.31 3.6
9/5/2006 1494/129 0.1 1.03 0.1 27894 3.31 0.5
9/13/2006 1494/171 0.2 1.02 0.1 26916 3.31 0
9/15/2006 1494/181 0.1 1.03 0.1 29553 3.29 0.2
9/15/2006 1494/187 0.1 1.12 0.1 32361 3.47 0.1
9/15/2006 1494/193 0.1 1.11 0.1 27303 3.12 0.2
9/15/2006 1494/199 0.2 1.02 0.1 29424 3.3 0
9/18/2006 1504/37 0.2 1.02 0.1 28020 3.27 0.2
9/18/2006 1504/42 0.6 1.03 0 27627 3.29 1.9
9/18/2006 1461/40 0.1 1.03 0.1 31109 3.66 0.2
10/5/2006 1504/65 0.1 1.04 0.1 36973 3.85 0.2
Average 0.1 1.04 0.1 29729 3.39 0.8
Min 0 1.02 0 26535 3.12 0
Max 0.6 1.12 0.2 37049 3.86 3.6
STDEV 0.128051 2770 0.168485904
3 Sigma 0.384153 8311 0.505457713
min -0.28 21418 2.88
max 0.48 38040 3.90
GAUSSIAN DISTRIBUTION
C.I. =
CONTROL CHART
SELECTIVITY AND SPECIFICITY
SelectivitySelectivity vs.vs. SpecificitySpecificity
SELECTIVITY AND SPECIFICITY
Selectivity:
A method’s ability to separate the analyte from other components
that may be present in the sample.
Definition of Selectivity from IUPAC: Selectivity of a method, refers to the
extent to which it can determine particular analytes under given conditions
in mixtures or matrices, simple or complex, without interferences from
other components.
SELECTIVITY AND SPECIFICITY
SELECTIVITY AND SPECIFICITY
Specificity:
A method’s ability to identify and measure absolutely and unequivocally the analyte
in the presence of the other components in the sample, such as; impurities,
degradation products, and excipients.
There must be inarguable supporting data for a method to be considered specific.
Specificity implies identification, purity tests, and assay (content or potency).
SELECTIVITY AND SPECIFICITY
Regulatory Requirements:
Stability indicating methods are not specified, but implied in 21 CFR Part 211.165
and 211.166 (3):
•211.165 (e) States that the accuracy, sensitivity, specificity, and reproducibility of
test methods employed by the firm shall be established and documented.
•211.166 (a) (3) Requires that test methods be reliable, meaningful, and specific.
STABILITY INDICATING METHOD (SIM) VS.
STABILITY SPECIFIC METHODS (SSM)
• Stability indicating assays accurately quantitate active ingredients
without interference from:
– Degradation products
– Process impurities
– Excipients
• A stability-specific method is one that meets all of the criteria above
but, in addition, the degradation components are detected and
quantitated.
Stress
Studies
“Absence of evidence is not evidence of absence”
- Carl Sagan,
The Dragons of Eden: Speculations on the Evolution of Human Intelligence
WHY DO WE PERFORM STRESS STUDIES?
Safety and Efficacy
Forced degradation or stress testing is undertaken to demonstrate specificity when
developing stability-indicating methods, particularly when little information is available
about potential degradation products.
WHY DO WE PERFORM STRESS STUDIES?
• Development and validation of stability-indicating methodology.
• Determination of degradation pathways of drug substances and drug products.
• Discernment of degradation products in formulations that are related to drug substances
versus those that are related to non-drug substances (excipients).
• Structure elucidation of degradation products.
• Determination of intrinsic stability of drug substance molecule.
WHY DO WE PERFORM STRESS STUDIES?
Defining characteristics of degradation studies:
• Carry out in solution and/or in the solid state.
• Involve conditions more severe than accelerated testing.
• Typically carry out on placebo, drug product, and API.
• Not part of formal stability program.
FORCED DEGRADATION (STRESS STUDIES)
Steps to Approaching Stress Studies in the Lab:
• Investigate the chemical structure and functional group.
• Study chemical and physical properties.
• Study synthetic route.
• Predict stress pathways based on storage conditions and
manufacturing process.
• Identify suitable separation method and detection.
• Design study based on the formulation (feed, tablet, ointment, etc.).
FORCED DEGRADATION (STRESS STUDIES)
Chemical Physical Environmental
Acid Agitation Heat
Base
Denaturation,
aggregation, adsorption
and precipitation
Light
(ICH Option I or II)
Oxidation RH
Deamidation Freeze/Thaw
Disulfide Bond Exchange
STRESS STUDY PATHWAYS
Pharmaceutical Biologics
Hydrolytic Hydrolytic
Oxidative Oxidative
Photolytic Aggregation
Thermolytic Deamidation
Disulfide Bond Exchange
FORCED DEGRADATION (STRESS STUDIES)
Stress Pathway Condition Time
Acid 0.01N 1 to 24 hours
Base 0.01N 1 o 24 hours
Oxidation 0.3% H2O2 1 to 24 hours
Light
600 to 800 foot candles
(sources include metal
halides, Hg, Xe lamp, or
UVB fluorescence)
Option II: 74Hours
Option I: 2-4 Hours
Heat/RH 40 °C/ 75% RH and
60 °C
24 to 72 hours
Freeze/Thaw -20 °C to 25 °C 3 Cycle of 24 hours
WHAT IS ADEQUATE STRESS?
Overstressing a molecule can lead to degradation profiles that are not
representative of primary degradation and are irrelevant to the stability of
the product.
Stress-testing conditions should be realistic, not excessive (5 – 10%).
FORCED DEGRADATION (STRESS STUDIES)
 Optimize detector setting
 Stress blank, placebo,
standard and sample
 Inject controls
 Extend run time
 Orthogonal Method
Overstress!!Overstress!!
EXAMPLE: PHOTOLYTIC STRESS
0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0 22.5 25.0 27.5 30.0
-2.20
0.00
1.25
2.50
3.80
1-SequenceName:ForcedDegradationLight SampleName:Fresh30mg/mLControl, Sample#12
2-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet1, Sample#17
3-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet2, Sample#26
4-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet3, Sample#35
mAU
min
4
3
2
1
Imp1-3.397
Imp3-6.097
Imp5-7.873
Imp6-8.320
DesMethyl-LVF-8.980
Levofloxacin-9.777
Imp7-10.733
Imp9-12.817
Imp10-13.320
Imp11-14.517
Imp14-20.487
WVL:280nm
EXAMPLE: ACID STRESSEXAMPLE: ACID STRESS
0.0 2.0 4.0 6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 22.0 24.0 26.0 28.0 30.0
-3.40
-2.00
0.00
2.60
1-SequenceName:ForcedDegradationAcid_Base SampleName:Fresh30mg/mLControl, Sample#17
2-SequenceName:ForcedDegradationAcid_Base SampleName:2hrsAcid30mg/mL, Sample#24
3-SequenceName:ForcedDegradationAcid_Base SampleName:4hrsAcid30mg/mL, Sample#38
4-SequenceName:ForcedDegradationAcid_Base SampleName:8hrsAcid30mg/mL, Sample#50
5-SequenceName:ForcedDegradationAcid_Base SampleName:24hrsAcid30mg/mL, Sample#64
mAU
min
5
4
3
2
1
Imp1-2.670
Imp3-3.387
Imp7-8.430
DesMethyl-LVF-9.097
Levofloxacin-9.890
WVL:280nm
EXAMPLE: HEAT STRESSEXAMPLE: HEAT STRESS
0.3 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0
-1.50
0.00
1.50
1-SequenceName:ForcedDegradationHeat SampleName:Fresh50mg/mLControl, Sample#17
2-SequenceName:ForcedDegradationHeat SampleName:4hrHeat,50mg/mLAssay, Sample#24
3-SequenceName:ForcedDegradationHeat SampleName:8hrHeat,50mg/mLAssay, Sample#31
4-SequenceName:ForcedDegradationHeat SampleName:24hrHeat,50mg/mLAssay, Sample#38
mAU
min
4
3
2
1
Imp1-2.660
Imp3-4.327
Imp4-6.070
Imp6-8.287
DesMethyl-LVF-8.960
Levofloxacin-9.753
WVL:280nm
MASS BALANCE1
From ICH Q1 A “Stability Testing of New Drug Substance and Product”
• The process of adding together the assay value and levels of degradation
products to see how closely these add up to 100 percent of the initial value,
with due consideration of the margin of analytical error1
.
MASS BALANCE
• Uncertainty in potency
• Loss of volatiles
• Diffusive losses
• Loss of UV chromophore
• Lack of universal detection
• Design of calculation
SOLUTION STABILITY
Purpose::
To determine stability of sample and standard Test solutions to support
duration of run sequence and potential investigation studies.
Procedure:
To evaluate several time intervals; (0, 24, 48, 72 hours), for both stock and
evaluated solution.
ESTABLISHING RANGE
• Range:
– Definition
– Criteria
• Limits of Detection and Quantitation
• Linearity
• Accuracy
• Precision
• Repeatability
DL & QL VERSUS SENSITIVITY
Sensitivity is measured by the slope of the calibration curve:Sensitivity is measured by the slope of the calibration curve:
 More sensitive method, steeper slope: Results in a larger change in the measured
response versus the controlled variable
DL & QL are measured by one of the four methods:DL & QL are measured by one of the four methods:
 lowest concentration for which RSD is <5.0%
 plot of standard deviation versus concentration
 95% CI of a best fit
 signal to noise ratio
DETERMINING DL AND QL:
Per ICH-Q2A:Per ICH-Q2A:
DL & QL can be calculated based on the standard deviation of the response (σ)
and the slope of the calibration curve (S) at levels approximating the limits
according to the following formulas:
DL= 3.3 (DL= 3.3 (σσ/S)/S)
QL= 10 (QL= 10 (σσ/S)/S)
The σ can be determined based on the σ of the blank, the residual σ of the
regression line, or the σ of y-intercepts of regression lines.
DETECTION LIMIT (DL)
4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0
-0.100
-0.000
0.100
0.200
0.300
1-LOD_LOQ#1 DiluentBlank UV_VIS_1
2-LOD_LOQ#11 LVFLOD(0.004ug/mL) UV_VIS_1
3-LOD_LOQ#12 LVFLOD(0.004ug/mL) UV_VIS_1
4-LOD_LOQ#13 LVFLOD(0.004ug/mL) UV_VIS_1
mAU
min
4
2-Levofloxacin-9.773
3
2-Levofloxacin-9.766
2
3-Levofloxacin-9.774
1
WVL:280nm
Quantitation Limit (QL)
4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0
-0.10
0.50
1.00
1.50
1-LOD_LOQ#1 DiluentBlank UV_VIS_1
2-LOD_LOQ#14 LVFLOQ(0.009ug/mL) UV_VIS_1
3-LOD_LOQ#15 LVFLOQ(0.009ug/mL) UV_VIS_1
4-LOD_LOQ#16 LVFLOQ(0.009ug/mL) UV_VIS_1
5-LOD_LOQ#17 LVFLOQ(0.009ug/mL) UV_VIS_1
6-LOD_LOQ#18 LVFLOQ(0.009ug/mL) UV_VIS_1
7-LOD_LOQ#19 LVFLOQ(0.009ug/mL) UV_VIS_1
mAU
min
7
2-Levofloxacin-9.767
6
2-Levofloxacin-9.771
5
2-Levofloxacin-9.789
4
3-Levofloxacin-9.781
3
3-Levofloxacin-9.778
2
2-Levofloxacin-9.772
1
WVL:280nm
LINEARITY
The ability of an analytical procedure (within a given range), to obtain test results
which are directly proportional to the concentration (amount) of analyte in the
sample.
LINEARITY CALCULATIONS
y = my = m••x + bx + b
Where: y = response, x = concentration, m = slope, and
b = y intercept
Percent Bias =
b
(x m) + b
100%
•
×
ACCURACY
• The measure of how close the experimental value is to the true value.
− Established across a specified range.
− Also called trueness.
ACCURACY
Determination of Accuracy:
• 9 determinations over 3 concentrations in triplicate preparation.
• The mean is an estimate of accuracy.
• RSD is an estimate of sample analysis precision.
ACCURACY
Should be reported as:
• The percent recovery by the assay of known added amount of analyte in the sample.
• The difference between the mean and the accepted true value together with the
confidence intervals.
DETERMINING ACCURACY FOR DRUG SUBSTANCES
• Use reference material.
• Compare procedure results with those of a second, well-characterized procedure.
• Infer from precision, linearity and specificity; 80,100 and 120% levels of label
claim.
DETERMINING ACCURACY FOR DRUG PRODUCTS
• Spike suitable Reference Materials into a Placebo.
• Add known quantities of analyte to the drug product.
• Compare procedure results with those of a second, well-characterized procedure.
• Infer from precision, linearity and specificity; 80, 100 and 120% levels of label claim.
DETERMINING ACCURACY FOR DRUG
PRODUCT RELATED SUBSTANCES
 Add known quantities of impurities to the sample.
Compare procedure results with those of a second, well-characterized procedure.
If impurities are not available, how do
you perform accuracy?
PRECISION
The closeness of agreement between a series of measurements, obtained
from a multiple sampling of the same homogeneous sample, under the
prescribed conditions.
PRECISION
Includes:Includes:
Repeatability
Intermediate Precision
Reproducibility
Report:Report:
Standard Deviation, Relative Standard
Deviation, Confidence Interval
REPRODUCIBILITY
• Expresses the precision between laboratories.
• Recommended parameters to be evaluated at the second laboratory include:
– Selectivity
– DL/QL
– Repeatability
– System Suitability
RUGGEDNESS
• Degree of reproducibility of test results under a variety of conditions:
−Different Laboratories
−Different Analysts
−Different Instruments
−Different Reagents
−Different Days
Ruggedness ≠ Robustness
MINIMUM SPECIFIED RANGES: DRUG SUBSTANCES
Impurity Reporting Thresholds
Maximum
Daily Dose
Qualification
and ID
Threshold
Reporting
Threshold
< 2g/day
0.1% or
1mg/day
(choose the
lower)
0.05%
> 2g/day 0.05% 0.03%
MINIMUM SPECIFIED RANGES: DRUG PRODUCTS
Degradation Product Reporting Thresholds In
New Drug Products:
Maximum Daily
Dose
Reporting
Threshold
≤ 1 g/day 0.1%
> 1 g/day 0.05%
ROBUSTNESS
• A measure of a method’s capacity to remain unaffected by small, deliberate variations in
method parameters.
• Provides an indication of a method’s reliability during normal usage.
• Assessed by making small, deliberate changes to the method and evaluating the results.
ROBUSTNESS
Examples of typical RP-HPLC variations:Examples of typical RP-HPLC variations:
pH of mobile phase
mobile phase composition
Ionic Strength
Different columns
Column temperature
flow rate
ROBUSTNESS
Parameter
Nominal
Procedure
Condition
Conditions Tested for
Robustness
Determination
MPA*-Buffer constituent pH 4.0 3.9, 4.1
MPA*-Buffer salt
concentration
10 mM Ammonium
Formate
9 mM, 11 mM
Column Temperature 30°C 25°C, 35°C
Detector Wavelength 290 nm 288 nm, 292 nm
Flow Rate 1.0 mL/min 0.9 mL/min, 1.1 mL/min
Injection Volume 20 µL 15 µL, 25 µL
*MPA = Mobile Phase A
METHOD REVALIDATION
Revalidate due to changes in:
Synthesis of the drug substance.
Composition of the drug product.
Analytical procedure.
ANALYTICAL METHOD LIFE CYCLE
Change to Method:
Evaluate the effect
Development of
the Method
Validation of the
Method
Method in
Routine use
Redevelopment of
the method required
due to change
Revalidation required
due to change
Change is not covered
by existing validation
Change is covered by
existing validation
REFERENCES
1. Bob Snider, CMC Group
2. ICH Q2 (R1)
3. Current USP <1224>
4. Current USP <1225>
5. Current USP <1226>
6. FDA Guidance for Industry
7. Miller, JM., Crowther, JB. 2000. Analytical Chemistry in a GMP
Environment. John Wiley & Sons, Inc.
WHAT IS SUCCESS?
Questions? Comments?
THANK YOU

More Related Content

What's hot

ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
Khadeeja6
 
Filter validation
Filter validationFilter validation
Filter validation
Sagar Savale
 
Computer system validations
Computer system validationsComputer system validations
Computer system validations
Amruta Balekundri
 
Stability testing protocols
Stability testing protocolsStability testing protocols
Stability testing protocols
MehulJain143
 
Cleaning and analytical validation
Cleaning and analytical validationCleaning and analytical validation
Cleaning and analytical validation
Malla Reddy College of Pharmacy
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
Sanathoiba Singha
 
Impurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek JainImpurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek Jain
Vivek Jain
 
Validation of utility system (water system)
Validation of utility system (water system)Validation of utility system (water system)
Validation of utility system (water system)
ShameerAbid
 
Analytical method validation, ICH Q2 guideline
Analytical method validation, ICH Q2 guidelineAnalytical method validation, ICH Q2 guideline
Analytical method validation, ICH Q2 guideline
Abhishek Soni
 
Qualification of gc equipment
Qualification of gc equipmentQualification of gc equipment
Qualification of gc equipment
srikrupa institute of pharmaceutical analysis
 
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
Peter van Amsterdam
 
Q3A(R2) IMPURITIES IN NEW DRUG SUBSTANCES
Q3A(R2)  IMPURITIES IN NEW DRUG SUBSTANCESQ3A(R2)  IMPURITIES IN NEW DRUG SUBSTANCES
Q3A(R2) IMPURITIES IN NEW DRUG SUBSTANCES
Muhamad Abdalkader
 
ANDA Stability Requirements
ANDA Stability RequirementsANDA Stability Requirements
ANDA Stability Requirements
GMP EDUCATION : Not for Profit Organization
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
Faris ameen
 
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological productsICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
saimsoleja
 
Process validation
Process validationProcess validation
Process validation
Saravanan subramaniyam
 
Anvisa gudelines
Anvisa gudelinesAnvisa gudelines
Anvisa gudelines
Ankit Gotecha
 
Stability Testing.pptx
Stability Testing.pptxStability Testing.pptx
Stability Testing.pptx
SunaynaChoudhary
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
akshara01
 
ICH Q6A Specifications by Chandra Mohan
ICH Q6A Specifications by Chandra MohanICH Q6A Specifications by Chandra Mohan
ICH Q6A Specifications by Chandra Mohan
Chandra Mohan
 

What's hot (20)

ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
ANALYSIS OF RAW MATERIALS, FINISHED PRODUCTS, PACKAGING MATERIALS, IPQC, CPCS...
 
Filter validation
Filter validationFilter validation
Filter validation
 
Computer system validations
Computer system validationsComputer system validations
Computer system validations
 
Stability testing protocols
Stability testing protocolsStability testing protocols
Stability testing protocols
 
Cleaning and analytical validation
Cleaning and analytical validationCleaning and analytical validation
Cleaning and analytical validation
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
 
Impurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek JainImpurities ICH Q3 Guidelines Au Vivek Jain
Impurities ICH Q3 Guidelines Au Vivek Jain
 
Validation of utility system (water system)
Validation of utility system (water system)Validation of utility system (water system)
Validation of utility system (water system)
 
Analytical method validation, ICH Q2 guideline
Analytical method validation, ICH Q2 guidelineAnalytical method validation, ICH Q2 guideline
Analytical method validation, ICH Q2 guideline
 
Qualification of gc equipment
Qualification of gc equipmentQualification of gc equipment
Qualification of gc equipment
 
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
The EMA Bioanalytical Method Validation Guideline: process, history, discussi...
 
Q3A(R2) IMPURITIES IN NEW DRUG SUBSTANCES
Q3A(R2)  IMPURITIES IN NEW DRUG SUBSTANCESQ3A(R2)  IMPURITIES IN NEW DRUG SUBSTANCES
Q3A(R2) IMPURITIES IN NEW DRUG SUBSTANCES
 
ANDA Stability Requirements
ANDA Stability RequirementsANDA Stability Requirements
ANDA Stability Requirements
 
Qualification of analytical instruments
Qualification of analytical instrumentsQualification of analytical instruments
Qualification of analytical instruments
 
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological productsICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
ICH q5c ICH Q5C Stability testing of Biotechnological / Biological products
 
Process validation
Process validationProcess validation
Process validation
 
Anvisa gudelines
Anvisa gudelinesAnvisa gudelines
Anvisa gudelines
 
Stability Testing.pptx
Stability Testing.pptxStability Testing.pptx
Stability Testing.pptx
 
cleaning validation..
cleaning validation..cleaning validation..
cleaning validation..
 
ICH Q6A Specifications by Chandra Mohan
ICH Q6A Specifications by Chandra MohanICH Q6A Specifications by Chandra Mohan
ICH Q6A Specifications by Chandra Mohan
 

Similar to Phase Appropriate Method Validation Aryo Boston-Nitto 2

Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
Stefan Holt
 
Seminar by prakash on validation
Seminar by prakash on validationSeminar by prakash on validation
Seminar by prakash on validation
prakash64742
 
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
 
Validation of Analytical method.ppt
Validation of Analytical method.pptValidation of Analytical method.ppt
Validation of Analytical method.ppt
Priyanka Yadav
 
HPLC validation.ppt
HPLC validation.pptHPLC validation.ppt
HPLC validation.ppt
Priyanka Yadav
 
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
 
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 validation
Analytical method validationAnalytical method validation
Analytical method validation
Malla Reddy College of Pharmacy
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
ceutics1315
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
Sai Praveen Reddy
 
Seminar on introduction to validation
Seminar on introduction to validationSeminar on introduction to validation
Seminar on introduction to validation
vasantipatil5
 
Method Validation - ICH /USP Validation, Linearity and Repeatability
Method Validation - ICH /USP Validation, Linearity and Repeatability Method Validation - ICH /USP Validation, Linearity and Repeatability
Method Validation - ICH /USP Validation, Linearity and Repeatability
labgo
 
Analytical quality by design
Analytical quality by designAnalytical quality by design
Analytical quality by design
Annalisa Forlenza
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
SUBHASISH DAS
 
PHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATIONPHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATION
JayeshRajput7
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
Gaurav Kr
 
Q2methodvalidation 161105173750
Q2methodvalidation 161105173750Q2methodvalidation 161105173750
Q2methodvalidation 161105173750
ChristianReckziegel1
 
ICH Q2 Analytical Method Validation
ICH Q2  Analytical Method ValidationICH Q2  Analytical Method Validation
ICH Q2 Analytical Method Validation
Naila Kanwal
 
Validation of Analytical Methods.pdf
Validation of Analytical Methods.pdfValidation of Analytical Methods.pdf
Validation of Analytical Methods.pdf
anjaneyulu49
 
ICH and WHO Guideline for Validation and Calibration.pptx
ICH and WHO Guideline for Validation and Calibration.pptxICH and WHO Guideline for Validation and Calibration.pptx
ICH and WHO Guideline for Validation and Calibration.pptx
Prachi Pandey
 

Similar to Phase Appropriate Method Validation Aryo Boston-Nitto 2 (20)

Analytical Method Validation
Analytical Method ValidationAnalytical Method Validation
Analytical Method Validation
 
Seminar by prakash on validation
Seminar by prakash on validationSeminar by prakash on validation
Seminar by prakash on validation
 
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
 
Validation of Analytical method.ppt
Validation of Analytical method.pptValidation of Analytical method.ppt
Validation of Analytical method.ppt
 
HPLC validation.ppt
HPLC validation.pptHPLC validation.ppt
HPLC validation.ppt
 
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
 
Life cycle of analytical method
Life cycle of analytical methodLife cycle of analytical method
Life cycle of analytical method
 
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
Analytical method validationAnalytical method validation
Analytical method validation
 
Seminar on introduction to validation
Seminar on introduction to validationSeminar on introduction to validation
Seminar on introduction to validation
 
Method Validation - ICH /USP Validation, Linearity and Repeatability
Method Validation - ICH /USP Validation, Linearity and Repeatability Method Validation - ICH /USP Validation, Linearity and Repeatability
Method Validation - ICH /USP Validation, Linearity and Repeatability
 
Analytical quality by design
Analytical quality by designAnalytical quality by design
Analytical quality by design
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
PHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATIONPHARMACEUTICAL VALIDATION
PHARMACEUTICAL VALIDATION
 
Analytical method validation
Analytical method validationAnalytical method validation
Analytical method validation
 
Q2methodvalidation 161105173750
Q2methodvalidation 161105173750Q2methodvalidation 161105173750
Q2methodvalidation 161105173750
 
ICH Q2 Analytical Method Validation
ICH Q2  Analytical Method ValidationICH Q2  Analytical Method Validation
ICH Q2 Analytical Method Validation
 
Validation of Analytical Methods.pdf
Validation of Analytical Methods.pdfValidation of Analytical Methods.pdf
Validation of Analytical Methods.pdf
 
ICH and WHO Guideline for Validation and Calibration.pptx
ICH and WHO Guideline for Validation and Calibration.pptxICH and WHO Guideline for Validation and Calibration.pptx
ICH and WHO Guideline for Validation and Calibration.pptx
 

Phase Appropriate Method Validation Aryo Boston-Nitto 2

  • 1. Phase Appropriate Method Validation Aryo Nikopour The Southern California Pharmaceutical Discussion Group (SCPDG) of AAPS Irvine, CA January 12, 2017
  • 6. PUBLISHED VALIDATION GUIDELINES  1978 Current Good Manufacturing Practices (cGMP)  1987 FDA Validation Guideline  1989 Supplement 9 to USP XXI  1994 CDER Reviewer Guidance: Validation of Chromatographic Method  1995 ICH Validation Definitions: Q2A, Text on Validation of Analytical procedures  1997 ICH Validation Methodology: Q2B, Validation of Analytical Procedures: Methodology  1999 Supplement 10 to USP 23 <1225>: Validation of Compendial Methods  1999 CDER “Bioanalytical Method Validation for Human Studies”  2000 CDER Draft “Analytical Procedures and Method Validation”  2014 CDER/CBER Guidance for Industry: “Analytical Procedure and Method Validation for Drug and Biologic”  PDA Technical Report No. 57 : Analytical Method Validation and Transfer for Biotechnology Products”
  • 7. GUIDELINES FOR METHOD VALIDATION www.ICH.orgwww.ICH.org (Dates indicate ICH finalization) ICH Q2(R1): Validation of Analytical Procedures: Methodology- Nov. 2005 ICH Q3A(R): Impurities in New Drug Substances - Feb. 2002 ICH Q3B(R): Impurities in New Drug Products – Feb. 2003 ICH Q3C: Impurities: Residual Solvents - July 1997 ICH Q5A,D: Biotech/Biological Products - 1997 ICH Q5B,C: Biotech/Biological Products - 1995
  • 8. GUIDELINES FOR METHOD VALIDATION www.ICH.orgwww.ICH.org (Dates indicate ICH finalization) ICH Q2(R1): Validation of Analytical Procedures: Methodology- Nov. 2005 ICH Q3A(R): Impurities in New Drug Substances - Feb. 2002 ICH Q3B(R): Impurities in New Drug Products – Feb. 2003 ICH Q3C: Impurities: Residual Solvents - July 1997 ICH Q5A,D: Biotech/Biological Products - 1997 ICH Q5B,C: Biotech/Biological Products - 1995
  • 9. VALIDATION IN THE 21 CENTURY
  • 11. CFR • There are many reason to validate analytical methods: – Regulatory Requirements – Good Science – Quality Control requirements. • The Code of Federal Regulations (CFR) 311.165c explicitly states that the, “Accuracy, Sensitivity, Specificity, and Reproducibility of test methods employed by the firm shall be established and documented.”
  • 12. ICH GUIDELINE Q2(R1) • The objective of validation of an analytical procedure is to demonstrate that it is suitable for its intended purpose, In practice, it is usually possible to design the experimental work such that the appropriate validation characteristics can be considered simultaneously, to provide a sound, overall knowledge of the capabilities of the analytical procedure, for instance; Specificity, Linearity, Range, Accuracy, and Precision. Support the identity, strength, quality, purity, and potency of the drug substances and drug products.
  • 13. WHAT IS METHOD VALIDATION?  Validation is procedure dependent.  Validation, “Proves” the procedure works as described.  Validation is product specific.  Procedures are instrument dependent.
  • 14. VERIFICATION USP <1226> • Current USP <1226> Verification of Compendial Procedure – The Analytical procedures in the current USP are legally recognized under section 501(b) of the Federal Food, Drug and Cosmetic Act as the regulatory analytical procedures for the compendial items. The suitability of these procedures must be verified under the actual conditions of use.
  • 15. VERIFICATION • When using USP analytical procedures, the guidance recommends that information be provided for the following characteristics: – Specificity of the procedure – Stability of the sample solution – Intermediate precision
  • 16. METHOD TRANSFER, USP <1224> • Method Transfer is a documented process that qualifies a laboratory (Receiving Lab) to use an analytical test procedure that is originated from the transferring laboratory. • Types of Method Transfer: – Comparative Testing – Co -validation – Revalidation/Partial Validation – Transfer Waiver
  • 17. CLASSIFICATION OF VALIDATED ANALYTICAL METHODS Compendial (USP 39/ NF 34): • Legally recognized under section 501 (b) of the Federal Food, Drug, and Cosmetic Act. • Recommends information be provided for; Specificity, Sample Solution Stability, and Intermediate Precision. Noncompendial: • Submitted with the NDA/ BLA or ANDA application. • If the compendial procedure is not stability-indicating, perform an alternative analytical procedure with complete validation.
  • 18. USP <1225>ASSAY CATEGORIES CategoryCategory NumberNumber Category NameCategory Name Description of AssayDescription of Assay I Quantitative Quantitation of major components/active ingredients present at high concentrations. II Impurities- Quantitative Determination of impurities or degradation products. II Impurities-Limit III Performance Characteristics Parameters to be tested depend on the nature of the test; includes dissolution testing. IV Identity
  • 19. METHOD VALIDATION REQUIREMENTS USP Assay Category I II III IV Parameter: Quantitative Limit Tests Accuracy Y Y • Y N Precision Y Y N Y N Intermediate Precision Y Y N Y N Specificity N Y Y N Y Detection Limit N Y Y • N Quantitation Limit N Y N • N Linearity Y Y N • N Range Y Y • • N Robustness Y Y N N N Selectivity Y Y N Y Y System Suitability Y Y N Y N Solution Stability Y Y N Y N • May be required, depending on the nature of the specific test.
  • 21. METHOD VALIDATION READINESS Define the application, purpose and scope of the method. Define Analytes, Dosage Strength and Sample Matrix. Review Method Development Summary Report. Evaluate method validation parameters during development.
  • 22. METHOD VALIDATION CHARACTERISTICS Validation Characteristics Experimental Details Acceptance Criteria Specificity Stress Studies 5-10% Degradation Selectivity Determine Chromatographic non- interference No inference , minimum resolution between peaks of interest and impurities should be >1.5 System Suitability System precision assessed by 6 replicate measurement/injections %RSD ≤2% Linearity At least 5 Concentration over the range Assay: 50% to 125% of Specification limit QL-150% of specification limit Calibration Model is valid R ≥0.998 Report Intercept, Slope and %Bias Detection Limit (DL) DL= 3.3 (DL= 3.3 (σσ/S)/S) S/N≥ 3S/N≥ 3 Quantitation Limit (QL) DL= 10 (DL= 10 (σσ/S)/S) %RSD≤ 15%%RSD≤ 15%
  • 23. METHOD VALIDATION CHARACTERISTICS Validation Characteristics Experimental Details Acceptance Criteria Precision : Repeatability Intermediate Precision (Ruggedness) Reproducibility 6 replicates 6 replicates %RSD≤ 2% Overall %RSD (two Analyst) Accuracy At least 9 determination over 3 concentration level e.g. 70 to 120% for For Assay Mean Recovery 97 to 103% for Impurities : 85% to 115% Range The range is defined by the results obtained for linearity, accuracy and precision Linearity, accuracy and precision demonstrated over the range Solution Stability Determine solution stability of Reference Standard Solution and Sample over 72 hours 98 to 102 % of control Robustness Deliberately change critical parameters of the method Must meet system suitability and selectivity requirements
  • 24. VALIDATION: PHASE I Drug Product Assay I.D. Quantitative Impurities Limit Test Selectivity X X X X Repeatability X X Accuracy/Precision Recovery at 100% At 100% of Reporting Threshold Linearity X QL to 200% of Limit Range Defined by ALP Defined by ALP DL/QL DL QL QL or at Limit System Suitability X X X X Solution Stability X X X
  • 25. VALIDATION: PHASE II Assay I.D Quantitative Impurities Limit Test Selectivity X X X X Specificity X Repeatability X X X Accuracy Recovery at 3 levels At 100% of Reporting Threshold Linearity X X X DL/QL DL X QL Range Define by ALP Defined by ALP System Suitability X X X X Solution Stability X X X
  • 26. VALIDATION: PHASE III Assay I.D Quantitative Impurities Limit Test Selectivity X X X X Specificity X Repeatability X X X Intermediate Precision X 2nd Analyst X X Accuracy X X Linearity X X DL/QL DL X QL Range Defined by ALP Defined by ALP Solution Stability X X X System Suitability X X X X Robustness X X X
  • 28. SYSTEM SUITABILITY Based on the concept that the equipment, electronics, analytical operations and samples to be analyzed constitute an integral system that can be evaluated as such. What parameters do you measure forWhat parameters do you measure for system suitabilitysystem suitability
  • 29. SYSTEM SUITABILITY What parameters do you measure for system suitability?What parameters do you measure for system suitability? CapacityCapacityEfficiencyEfficiencySelectivitySelectivity
  • 30. Date NB/Page Standard B1 (n=6) Injections SST Solution B1 and B2 Area %RSD Average Tailing Factor %RSD Retention Time Average Theoretical Plates Resolution LVF and DesMethyl-LVF) Response Factor % Difference ≤ 1% 0.8 ≤ Tf ≤ 1.4 ≤ 1% >15000 NLT 2.5 ≤ 2% 8/18/2006 1494/18 0.1 1.03 0.1 29834 3.27 0.2 8/18/2006 1494/31 0.1 1.03 0 32177 3.28 0.1 8/20/2006 1494/52 0.1 1.03 0.1 27792 3.3 0.7 8/22/2006 1494/72 0.1 1.03 0 26567 3.31 2.7 8/23/2006 1504/1 0 1.03 0.2 27228 3.29 0.8 8/24/2006 1504/8 0.2 1.02 0.1 26535 3.32 1.2 8/25/2006 1504/17 0.1 1.02 0.1 26903 3.31 3.6 9/5/2006 1494/129 0.1 1.03 0.1 27894 3.31 0.5 9/13/2006 1494/171 0.2 1.02 0.1 26916 3.31 0 9/15/2006 1494/181 0.1 1.03 0.1 29553 3.29 0.2 9/15/2006 1494/187 0.1 1.12 0.1 32361 3.47 0.1 9/15/2006 1494/193 0.1 1.11 0.1 27303 3.12 0.2 9/15/2006 1494/199 0.2 1.02 0.1 29424 3.3 0 9/18/2006 1504/37 0.2 1.02 0.1 28020 3.27 0.2 9/18/2006 1504/42 0.6 1.03 0 27627 3.29 1.9 9/18/2006 1461/40 0.1 1.03 0.1 31109 3.66 0.2 10/5/2006 1504/65 0.1 1.04 0.1 36973 3.85 0.2 Average 0.1 1.04 0.1 29729 3.39 0.8 Min 0 1.02 0 26535 3.12 0 Max 0.6 1.12 0.2 37049 3.86 3.6 STDEV 0.128051 2770 0.168485904 3 Sigma 0.384153 8311 0.505457713 min -0.28 21418 2.88 max 0.48 38040 3.90
  • 33. SELECTIVITY AND SPECIFICITY SelectivitySelectivity vs.vs. SpecificitySpecificity
  • 34. SELECTIVITY AND SPECIFICITY Selectivity: A method’s ability to separate the analyte from other components that may be present in the sample. Definition of Selectivity from IUPAC: Selectivity of a method, refers to the extent to which it can determine particular analytes under given conditions in mixtures or matrices, simple or complex, without interferences from other components.
  • 36. SELECTIVITY AND SPECIFICITY Specificity: A method’s ability to identify and measure absolutely and unequivocally the analyte in the presence of the other components in the sample, such as; impurities, degradation products, and excipients. There must be inarguable supporting data for a method to be considered specific. Specificity implies identification, purity tests, and assay (content or potency).
  • 37. SELECTIVITY AND SPECIFICITY Regulatory Requirements: Stability indicating methods are not specified, but implied in 21 CFR Part 211.165 and 211.166 (3): •211.165 (e) States that the accuracy, sensitivity, specificity, and reproducibility of test methods employed by the firm shall be established and documented. •211.166 (a) (3) Requires that test methods be reliable, meaningful, and specific.
  • 38. STABILITY INDICATING METHOD (SIM) VS. STABILITY SPECIFIC METHODS (SSM) • Stability indicating assays accurately quantitate active ingredients without interference from: – Degradation products – Process impurities – Excipients • A stability-specific method is one that meets all of the criteria above but, in addition, the degradation components are detected and quantitated.
  • 39. Stress Studies “Absence of evidence is not evidence of absence” - Carl Sagan, The Dragons of Eden: Speculations on the Evolution of Human Intelligence
  • 40. WHY DO WE PERFORM STRESS STUDIES? Safety and Efficacy Forced degradation or stress testing is undertaken to demonstrate specificity when developing stability-indicating methods, particularly when little information is available about potential degradation products.
  • 41. WHY DO WE PERFORM STRESS STUDIES? • Development and validation of stability-indicating methodology. • Determination of degradation pathways of drug substances and drug products. • Discernment of degradation products in formulations that are related to drug substances versus those that are related to non-drug substances (excipients). • Structure elucidation of degradation products. • Determination of intrinsic stability of drug substance molecule.
  • 42. WHY DO WE PERFORM STRESS STUDIES? Defining characteristics of degradation studies: • Carry out in solution and/or in the solid state. • Involve conditions more severe than accelerated testing. • Typically carry out on placebo, drug product, and API. • Not part of formal stability program.
  • 43. FORCED DEGRADATION (STRESS STUDIES) Steps to Approaching Stress Studies in the Lab: • Investigate the chemical structure and functional group. • Study chemical and physical properties. • Study synthetic route. • Predict stress pathways based on storage conditions and manufacturing process. • Identify suitable separation method and detection. • Design study based on the formulation (feed, tablet, ointment, etc.).
  • 44. FORCED DEGRADATION (STRESS STUDIES) Chemical Physical Environmental Acid Agitation Heat Base Denaturation, aggregation, adsorption and precipitation Light (ICH Option I or II) Oxidation RH Deamidation Freeze/Thaw Disulfide Bond Exchange
  • 45. STRESS STUDY PATHWAYS Pharmaceutical Biologics Hydrolytic Hydrolytic Oxidative Oxidative Photolytic Aggregation Thermolytic Deamidation Disulfide Bond Exchange
  • 46. FORCED DEGRADATION (STRESS STUDIES) Stress Pathway Condition Time Acid 0.01N 1 to 24 hours Base 0.01N 1 o 24 hours Oxidation 0.3% H2O2 1 to 24 hours Light 600 to 800 foot candles (sources include metal halides, Hg, Xe lamp, or UVB fluorescence) Option II: 74Hours Option I: 2-4 Hours Heat/RH 40 °C/ 75% RH and 60 °C 24 to 72 hours Freeze/Thaw -20 °C to 25 °C 3 Cycle of 24 hours
  • 47. WHAT IS ADEQUATE STRESS? Overstressing a molecule can lead to degradation profiles that are not representative of primary degradation and are irrelevant to the stability of the product. Stress-testing conditions should be realistic, not excessive (5 – 10%).
  • 48. FORCED DEGRADATION (STRESS STUDIES)  Optimize detector setting  Stress blank, placebo, standard and sample  Inject controls  Extend run time  Orthogonal Method Overstress!!Overstress!!
  • 49. EXAMPLE: PHOTOLYTIC STRESS 0.0 2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0 22.5 25.0 27.5 30.0 -2.20 0.00 1.25 2.50 3.80 1-SequenceName:ForcedDegradationLight SampleName:Fresh30mg/mLControl, Sample#12 2-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet1, Sample#17 3-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet2, Sample#26 4-SequenceName:ForcedDegradationLight SampleName:LightStressed30mg/mLSet3, Sample#35 mAU min 4 3 2 1 Imp1-3.397 Imp3-6.097 Imp5-7.873 Imp6-8.320 DesMethyl-LVF-8.980 Levofloxacin-9.777 Imp7-10.733 Imp9-12.817 Imp10-13.320 Imp11-14.517 Imp14-20.487 WVL:280nm
  • 50. EXAMPLE: ACID STRESSEXAMPLE: ACID STRESS 0.0 2.0 4.0 6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 22.0 24.0 26.0 28.0 30.0 -3.40 -2.00 0.00 2.60 1-SequenceName:ForcedDegradationAcid_Base SampleName:Fresh30mg/mLControl, Sample#17 2-SequenceName:ForcedDegradationAcid_Base SampleName:2hrsAcid30mg/mL, Sample#24 3-SequenceName:ForcedDegradationAcid_Base SampleName:4hrsAcid30mg/mL, Sample#38 4-SequenceName:ForcedDegradationAcid_Base SampleName:8hrsAcid30mg/mL, Sample#50 5-SequenceName:ForcedDegradationAcid_Base SampleName:24hrsAcid30mg/mL, Sample#64 mAU min 5 4 3 2 1 Imp1-2.670 Imp3-3.387 Imp7-8.430 DesMethyl-LVF-9.097 Levofloxacin-9.890 WVL:280nm
  • 51. EXAMPLE: HEAT STRESSEXAMPLE: HEAT STRESS 0.3 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0 -1.50 0.00 1.50 1-SequenceName:ForcedDegradationHeat SampleName:Fresh50mg/mLControl, Sample#17 2-SequenceName:ForcedDegradationHeat SampleName:4hrHeat,50mg/mLAssay, Sample#24 3-SequenceName:ForcedDegradationHeat SampleName:8hrHeat,50mg/mLAssay, Sample#31 4-SequenceName:ForcedDegradationHeat SampleName:24hrHeat,50mg/mLAssay, Sample#38 mAU min 4 3 2 1 Imp1-2.660 Imp3-4.327 Imp4-6.070 Imp6-8.287 DesMethyl-LVF-8.960 Levofloxacin-9.753 WVL:280nm
  • 52. MASS BALANCE1 From ICH Q1 A “Stability Testing of New Drug Substance and Product” • The process of adding together the assay value and levels of degradation products to see how closely these add up to 100 percent of the initial value, with due consideration of the margin of analytical error1 .
  • 53. MASS BALANCE • Uncertainty in potency • Loss of volatiles • Diffusive losses • Loss of UV chromophore • Lack of universal detection • Design of calculation
  • 54. SOLUTION STABILITY Purpose:: To determine stability of sample and standard Test solutions to support duration of run sequence and potential investigation studies. Procedure: To evaluate several time intervals; (0, 24, 48, 72 hours), for both stock and evaluated solution.
  • 55. ESTABLISHING RANGE • Range: – Definition – Criteria • Limits of Detection and Quantitation • Linearity • Accuracy • Precision • Repeatability
  • 56. DL & QL VERSUS SENSITIVITY Sensitivity is measured by the slope of the calibration curve:Sensitivity is measured by the slope of the calibration curve:  More sensitive method, steeper slope: Results in a larger change in the measured response versus the controlled variable DL & QL are measured by one of the four methods:DL & QL are measured by one of the four methods:  lowest concentration for which RSD is <5.0%  plot of standard deviation versus concentration  95% CI of a best fit  signal to noise ratio
  • 57. DETERMINING DL AND QL: Per ICH-Q2A:Per ICH-Q2A: DL & QL can be calculated based on the standard deviation of the response (σ) and the slope of the calibration curve (S) at levels approximating the limits according to the following formulas: DL= 3.3 (DL= 3.3 (σσ/S)/S) QL= 10 (QL= 10 (σσ/S)/S) The σ can be determined based on the σ of the blank, the residual σ of the regression line, or the σ of y-intercepts of regression lines.
  • 58. DETECTION LIMIT (DL) 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0 -0.100 -0.000 0.100 0.200 0.300 1-LOD_LOQ#1 DiluentBlank UV_VIS_1 2-LOD_LOQ#11 LVFLOD(0.004ug/mL) UV_VIS_1 3-LOD_LOQ#12 LVFLOD(0.004ug/mL) UV_VIS_1 4-LOD_LOQ#13 LVFLOD(0.004ug/mL) UV_VIS_1 mAU min 4 2-Levofloxacin-9.773 3 2-Levofloxacin-9.766 2 3-Levofloxacin-9.774 1 WVL:280nm
  • 59. Quantitation Limit (QL) 4.0 5.0 6.0 7.0 8.0 9.0 10.0 11.0 12.0 13.0 14.0 15.0 -0.10 0.50 1.00 1.50 1-LOD_LOQ#1 DiluentBlank UV_VIS_1 2-LOD_LOQ#14 LVFLOQ(0.009ug/mL) UV_VIS_1 3-LOD_LOQ#15 LVFLOQ(0.009ug/mL) UV_VIS_1 4-LOD_LOQ#16 LVFLOQ(0.009ug/mL) UV_VIS_1 5-LOD_LOQ#17 LVFLOQ(0.009ug/mL) UV_VIS_1 6-LOD_LOQ#18 LVFLOQ(0.009ug/mL) UV_VIS_1 7-LOD_LOQ#19 LVFLOQ(0.009ug/mL) UV_VIS_1 mAU min 7 2-Levofloxacin-9.767 6 2-Levofloxacin-9.771 5 2-Levofloxacin-9.789 4 3-Levofloxacin-9.781 3 3-Levofloxacin-9.778 2 2-Levofloxacin-9.772 1 WVL:280nm
  • 60. LINEARITY The ability of an analytical procedure (within a given range), to obtain test results which are directly proportional to the concentration (amount) of analyte in the sample.
  • 61. LINEARITY CALCULATIONS y = my = m••x + bx + b Where: y = response, x = concentration, m = slope, and b = y intercept Percent Bias = b (x m) + b 100% • ×
  • 62. ACCURACY • The measure of how close the experimental value is to the true value. − Established across a specified range. − Also called trueness.
  • 63. ACCURACY Determination of Accuracy: • 9 determinations over 3 concentrations in triplicate preparation. • The mean is an estimate of accuracy. • RSD is an estimate of sample analysis precision.
  • 64. ACCURACY Should be reported as: • The percent recovery by the assay of known added amount of analyte in the sample. • The difference between the mean and the accepted true value together with the confidence intervals.
  • 65. DETERMINING ACCURACY FOR DRUG SUBSTANCES • Use reference material. • Compare procedure results with those of a second, well-characterized procedure. • Infer from precision, linearity and specificity; 80,100 and 120% levels of label claim.
  • 66. DETERMINING ACCURACY FOR DRUG PRODUCTS • Spike suitable Reference Materials into a Placebo. • Add known quantities of analyte to the drug product. • Compare procedure results with those of a second, well-characterized procedure. • Infer from precision, linearity and specificity; 80, 100 and 120% levels of label claim.
  • 67. DETERMINING ACCURACY FOR DRUG PRODUCT RELATED SUBSTANCES  Add known quantities of impurities to the sample. Compare procedure results with those of a second, well-characterized procedure. If impurities are not available, how do you perform accuracy?
  • 68. PRECISION The closeness of agreement between a series of measurements, obtained from a multiple sampling of the same homogeneous sample, under the prescribed conditions.
  • 70. REPRODUCIBILITY • Expresses the precision between laboratories. • Recommended parameters to be evaluated at the second laboratory include: – Selectivity – DL/QL – Repeatability – System Suitability
  • 71. RUGGEDNESS • Degree of reproducibility of test results under a variety of conditions: −Different Laboratories −Different Analysts −Different Instruments −Different Reagents −Different Days Ruggedness ≠ Robustness
  • 72. MINIMUM SPECIFIED RANGES: DRUG SUBSTANCES Impurity Reporting Thresholds Maximum Daily Dose Qualification and ID Threshold Reporting Threshold < 2g/day 0.1% or 1mg/day (choose the lower) 0.05% > 2g/day 0.05% 0.03%
  • 73. MINIMUM SPECIFIED RANGES: DRUG PRODUCTS Degradation Product Reporting Thresholds In New Drug Products: Maximum Daily Dose Reporting Threshold ≤ 1 g/day 0.1% > 1 g/day 0.05%
  • 74. ROBUSTNESS • A measure of a method’s capacity to remain unaffected by small, deliberate variations in method parameters. • Provides an indication of a method’s reliability during normal usage. • Assessed by making small, deliberate changes to the method and evaluating the results.
  • 75. ROBUSTNESS Examples of typical RP-HPLC variations:Examples of typical RP-HPLC variations: pH of mobile phase mobile phase composition Ionic Strength Different columns Column temperature flow rate
  • 76. ROBUSTNESS Parameter Nominal Procedure Condition Conditions Tested for Robustness Determination MPA*-Buffer constituent pH 4.0 3.9, 4.1 MPA*-Buffer salt concentration 10 mM Ammonium Formate 9 mM, 11 mM Column Temperature 30°C 25°C, 35°C Detector Wavelength 290 nm 288 nm, 292 nm Flow Rate 1.0 mL/min 0.9 mL/min, 1.1 mL/min Injection Volume 20 µL 15 µL, 25 µL *MPA = Mobile Phase A
  • 77. METHOD REVALIDATION Revalidate due to changes in: Synthesis of the drug substance. Composition of the drug product. Analytical procedure.
  • 78. ANALYTICAL METHOD LIFE CYCLE Change to Method: Evaluate the effect Development of the Method Validation of the Method Method in Routine use Redevelopment of the method required due to change Revalidation required due to change Change is not covered by existing validation Change is covered by existing validation
  • 79. REFERENCES 1. Bob Snider, CMC Group 2. ICH Q2 (R1) 3. Current USP <1224> 4. Current USP <1225> 5. Current USP <1226> 6. FDA Guidance for Industry 7. Miller, JM., Crowther, JB. 2000. Analytical Chemistry in a GMP Environment. John Wiley & Sons, Inc.