You are on page 1of 102

Compendial HPLC Practices

Behnam Davani
USP Consultant

Last Update: January 2018


Horacio Pappa
CM-621-02

Disclaimer

Because USP text and publications may have legal implications in the U.S. and elsewhere, their
language must stand on its own. The USP shall not provide an official ex post facto interpretation
to one party, thereby placing other parties without that interpretation at a possible disadvantage.
The requirements shall be uniformly and equally available to all parties.

In addition, USP shall not provide an official opinion as to whether a particular article does or does
not comply with compendial requirements, except as part of an established USP verification or
other conformity assessment program that is conducted separately from and independent of USP's
standard-setting activities.

Certain commercial equipment, instruments or materials may be identified in this presentation to


specify adequately the experimental procedure. Such identification does not imply approval,
endorsement, or certification by USP of a particular brand or product, nor does it imply that the
equipment, instrument or material is necessarily the best available for the purpose or that any
other brand or product was judged to be unsatisfactory or inadequate.

2
© 2018 USP
Behnam Davani

USP Affiliation: USP Consultant (Former USP employee 1999 – July 2017)
Title: Consultant
Company: Independent Consultant
Education: Ph.D. in Analytical Chemistry
MBA in Management

Dr. Behnam Davani is currently an Independent Consultant. He has more than 28 years of experience in industry,
Instructor Photo compendial/regulatory science and training. He has extensive experience as faculty for the development and
teaching of several compendial and cGMP courses both domestically and internationally for USP Global Education
Program. Dr. Davani is also editor and author of a recent book: "Pharmaceutical Analysis for Small Molecules",
published in August 2017.

Prior to this position, he was the Principal Scientific Liaison between the USP Science Division and the USP
Education Department. He was responsible for scientific management of the development and teaching of USP
Education courses for stakeholders worldwide. Additional responsibilities included:
• Faculty for several Pharmacopeial Education course topics worldwide (China, India, Russia, Korea, Europe, Latin America and Middle East).
These courses included validation/verification/transfer of analytical procedures, impurities in drug substances and drug products, compendial
HPLC, residual solvents, spectroscopy and stability studies for pharmaceutical products.
• Providing scientific support to the USP International Regulatory Affairs Department and Global Public Health Department for outreach and
training for international regulatory agencies.
Prior to this position, Dr. Davani was Director of the Chemical Medicine Department at USP. In this capacity, he provided scientific leadership and
training to a team of US and internationally based staff, responsible for the development and modernization of USP monographs for low molecular
weight pharmaceuticals
Prior to joining USP in 1999, Dr. Davani worked in industry (Sigma-Aldrich, MRI Global) for more than ten years in various technical management
positions. He is a member of the American Chemical Society and the American Association of Pharmaceutical Scientists.

3
© 2018 USP

Outline

 Theory
 Equipment
 System Suitability
 Adjustments to the Chromatographic System
 Harmonization
 Validation of Compendial Procedures

4
© 2018 USP
Compendial HPLC Practices
Theory

Chromatography

 Chromatography is a separation technique


 Components of a mixture are distributed between two phases, one is stationary
and the other mobile.
 The stationary phase may be a solid or a liquid supported on a solid.
 The mobile phase may be gaseous or liquid or supercritical fluid.
 The separation may be based on adsorption, partition, or ion exchange; or it
may be based on differences in molecules, size, mass, or volume.

USP 36–NF 31 <621> 6


© 2018 USP
Chromatography

 Types of Chromatographic tests in USP


– Paper--useful for identification, convenient and simple
– Thin-layer--useful for identification, convenient and simple
– Column--useful for the separation of individual compounds from mixtures
– Gas--requires more elaborate instrumentation, useful for identification and quantitation
including low level impurities
– HPLC--requires more elaborate instrumentation, useful for identification and quantitation
including low level impurities

7
© 2018 USP

HPLC

 HPLC-High Performance (Pressure) Liquid Chromatography


– Definition: Separation Technique based on a solid stationary phase and a liquid mobile phase
– Separation Mechanisms: partition, adsorption, ion-exchange or molecular size depending on
the type of stationary phase used
– Useful for drugs, nonvolatile or thermally unstable compounds

8
© 2018 USP
Solvent Polarities

 Common solvents in order of increasing polarity:


– Pentane
– Hexane, Heptane, Isooctane
– Toluene
– Isopropyl alcohol
– Tetrahydrofuran
– Chloroform
– Acetone, methanol
– Acetonitrile
– Water
Reference: L.R. Snyder, J. Chromatogr., 92, 223 (1974); J Chromatogr. Sci., 16, 223 (1978)
9
© 2018 USP

Normal Phase HPLC

 Polar stationary phase and a non-polar mobile phase


 Typical column: L3
 Typical mobile phase: n-Hexane

 Example: Anthralin Assay


 Column: L3
 Mobile Phase: n-hexane, dichloromethane, and glacial acetic acid (82: 12: 6)

10
© 2018 USP
Reversed Phase HPLC

 Non polar stationary phase with a polar mobile phase


 Typical columns used are L1 (C18) and L7 (C8)
 Typical mobile phase include a mixture of aqueous buffer with an organic
solvent or water and an organic solvent

 Example: Acetaminophen Capsules Assay


 Column: L1
 Mobile Phase: water and methanol (3:1)

11
© 2018 USP

Ion Pair Chromatography

 Ionized or ionizable sample components (charged)


 Ion Pair reagent (opposite charge) add to the mobile phase to form neutral
complex (ion-pair) with the sample component
 Separate using a reversed phase column
 The more hydrophobic the pairing agent the greater the retention on the column

Solute RP Ion-Paring Reagent

Anionic (Acid) Tetraalkylammonium cation

Cationic (Base) Alkyl Sulfonate Anion

12
© 2018 USP
Ion Pair Chromatography

 Example: Tripelennamine Hydrochloride


 Column: L7
 Mobile Phase: 530 mL methanol, 1.0 mL N,N-dimethyloctylamine, 430 mL of
Ion-pair solution adjust to
pH 3.0
 Ion-pair solution: 29 mM sodium 1-octanesulfonate solution

13
© 2018 USP

Ion-Exchange Chromatography

 Stationary phase synthetic organic resins


 Separation affected by pH, temperature, ionic concentration, organic modifiers,
etc.
 Increasing the ionic concentration causes competition for active sites which
displaces solutes
 Vary pH to get differences in the charge of various sample components. Charge
depends on pH of solvent and pKa of ionizable group
 Useful for peptides, proteins, oligonucleotides, carbohydrates, etc.

14
© 2018 USP
Ion-Exchange Chromatography

 Cation exchange resins contain negatively charged active sites to separate


basic substances like amines
 Typical columns used L6, L17, L22, L19, L34

 Example: Lactitol Assay


 Column: L34
 Mobile Phase: water

15
© 2018 USP

Ion-Exchange Chromatography

 Anion exchange resins contain positively charged active sites for separation of
negatively charged groups such as phosphate and carboxylate
 Typical columns used are L12, L23, L31

 Example: Cinoxacin Assay


 Column: L12
 Mobile Phase: Dilute 100.0 mL of Sodium borate solution and 0.426 g of sodium
sulfate with water to 1000mL

16
© 2018 USP
Size Exclusion Chromatography

 Gel or Porous stationary phase


 Separation by size
 Solute exchange between mobile phase in and outside of pores
 Too large to enter pores = unretained
 Increased retention with smaller size molecules
 Pore size range of stationary phase determines size range of separated
components

17
© 2018 USP

Size Exclusion Chromatography

 Gel permeation chromatography: nonaqueous mobile phase


 Gel filtration chromatography: aqueous mobile phase
 Typical columns used L25, L33, L37, L54

 Example: Dextran 1 Molecular weight distribution and average molecular weight


 Column: L54
 Mobile Phase: sodium chloride solution 2.9 g/L

18
© 2018 USP
Hydrodynamics of Chromatography

 Peak width is dictated by the HETP (Height Equivalent to a Theoretical Plate)

 HETP will depends on the mobile phase hydrodynamics, the stationary phase
and the properties of the mixture to be separated

19
© 2018 USP

The Van Deemter Equation

H= A · dp + B/µ + C · de2 · µ

Eddy Diffusion
Longitudinal Diffusion
Multi-path Effect Mass Transfer (Kinetics)
dp = Diameter of particle
µ = Linear velocity de = “Effective particle
Also related to the quality size” (In the case of fully
of the packing of the porous particles this is dp)
column

20
© 2018 USP
Van Deemter: “A” Term

H = A·dp + B/µ + C·de2·µ


Eddy Diffusion or Multi-Path-Effect

Desired:
• Smaller particles
• Narrow particle size distribution
with uniform shape
• Optimized packing

Dispersion due to eddy diffusion or multi-path effect

21
© 2018 USP

Van Deemter: “B” Term

H= A · dp + B/µ + C · de2 · µ
Longitudinal Diffusion

For HPLC
Longitudinal diffusion inside the Zero linear velocity
column is negligible with higher
linear velocities and independent of
w1/2
particle size
6

Very low linear velocity tR

w1/2
5 6

At or above optimum linear velocity tR


22
© 2018 USP
Van Deemter: “C” Term

H = A·dp + B/µ + C·de2·µ


Mass Transfer Kinetics

23
© 2018 USP

The Van Deemter Equation

H= A · dp + B/µ + C · de2 · µ

Eddy Diffusion
Longitudinal Diffusion
Multi-path Effect Mass Transfer (Kinetics)
dp = Diameter of particle
µ = Linear velocity de = “Effective particle
Also related to the quality size” (In the case of fully
of the packing of the porous particles this is dp)
column
Plate Height (H)

C
B
A
Mobile phase velocity (µ)
24
© 2018 USP
Resolution Equation

N  α - 1  k 
RS = •  • 
4  α   1 + k 
Efficiency Selectivity Retention

N = Theoretical plates
α = t’R2/t’R1
k = tR-to/to

25
© 2018 USP

Retention

N  α - 1  k 
RS = •  • 1 + k 
4  α   
Efficiency Selectivity Retention
Term Term Term

 Varies with mobile phase composition

 Optimal 2 ≤ k ≤ 10

26
© 2018 USP
Selectivity

 Related to peak separation

 Depends on stationary phase, solvent, temperature, etc.


High α

N  α - 1  k 
RS = • •
4  α   1 + k 

Efficiency Selectivity Retention
Term Term Term Low α

27
© 2018 USP

Efficiency

Low N
N  α - 1  k 
RS = •  • 1 + k 
4  α   
Efficiency Selectivity Retention
Term Term Term
High N

 It dictates the peak width


 Depends mainly on the column length and particle size

28
© 2018 USP
Compendial HPLC Practices
Equipment
HPLC Equipment Components

 Pump
 Injector
 Column
 Detector
 Data Collection/Analysis Device

31
© 2018 USP

Pumping Systems

 Deliver Mobile phase at a constant rate with minimal fluctuations


 Operating pressure up to 6000 psi (400 bar)
 Inert to corrosive mobile phase components
 Programmable to vary the range of Mobile phase components--Gradient or
Isocratic Mixing
– High pressure mixing
– Low pressure mixing
 Most common-dual piston reciprocating pump

32
© 2018 USP
Pumping Systems

 Isocratic run--the mobile phase composition is kept the same throughout the run
– Pre-mix the mobile phase
– Mix mobile phase by pump

 Gradient run--changing the mobile phase composition during the


chromatographic run
 Gradient useful for complex mixtures with different capacity factors to reduce
analysis time

33
© 2018 USP

High Pressure Mixing System

34
© 2018 USP
Low Pressure Mixing System

35
© 2018 USP

Dead Volume, Dwell Volume, etc.

 Dead Volume: Volume of mobile phase inside the column


 Extra-column Volume: All the volume accessible to the analyte outside the
column
 Dwell Volume: Only apply to gradients. Represents the volume of mobile phase
from the point where the gradient is formed to the column inlet. It is critical
during method transfer

36
© 2018 USP
Dead Volume, Dwell Volume, etc.

Dwell volume determination (Eu. Ph.)

• Gradient: Water / 0.1% acetone in water


• Gradient duration (tG) 20’
• Detection 265 nm
• Without column

tD

tD=t0.5 – 0.5tG

Dwell Volume (D)= tD x F


37
© 2018 USP

Injector

 Dissolved solute is injected into the mobile phase


 Manually
– Six port two position valve.
– Position 1 flow moves from pump to column and bypasses injector loop
– Position 2 flow moves from pump through injector loop to column

 Automatically by autosampler
– Useful for quality control and high throughput screening
– May need to change syringe or loop based on injection amount
– May need to check precision and linearity, sample carryover, wash solvent, vial filling-minimum
and maximum
38
© 2018 USP
Injector

 Want sample to enter column as a tight band or plug


 Avoid excess tubing (length and diameter) between injector and column and
between column and detector and connectors with dead volume which may
cause the sample band to become broader before entering the column
 Band broadening increases peak widths which decreases column efficiency and
decreases resolution
 Band broadening can also occur as the sample passes through the column

39
© 2018 USP

Columns

Monograph specifies packing category: L1, L2, L3…


Reagents, Indicators, and Solutions—Chromatographic Columns defines
packing categories

40
© 2018 USP
Columns (cont)

Different brands of columns in the same category may behave differently in a


particular method:
– Behavior depends on:
• Carbon load
• Surface area
• Endcapping
• Particle shape
• Particle size
• Effect of surface silanols

Variations may also occur with different lots of the same brand

41
© 2018 USP

Solid Support Types

 Silica
– Type A
– Type B

 Polymers

 Hybrids

42
© 2018 USP
Silica Materials

 Structurally strong

 Easy-to-modify surface

 pH stability (pH 1.5 – 8 or 10)

 High efficiency

43
© 2018 USP

Type A Silica

 Old type (Irregular or spherical)

– Low amount of bonded or associated silanols

– High metal content (>1000 ppm)

• Metals activate silanols, inducing ionization

• Metals chelate acids, interact ionically with bases

44
© 2018 USP
Type B Silica

 Newer Type

– High amount of bonded or associated silanols

– Ultra-high purity (>99.9%)

– Low metal content (<50 ppm)

– Better peak shape for acids and bases

45
© 2018 USP

Silica Bonded Phase – pH stability

Low pH Ligand Hydrolysis High pH Silica Dissolution

H+
H+ OH- OH-
H+ OH-
H H
O O O O
O Si O Si O O Si O Si O

O O O O

Silica Surface w/ Alkyl Chain Silica Surface w/ Alkyl Chain

46
© 2018 USP
Polymeric Particles

 Extended pH stability (1-14)


 Chemically inert
 Poor mechanical strength
– Lower efficiency
– Less reproducible

 Very hydrophobic

47
© 2018 USP

Efficiency Comparison - Polymer vs. Silica

Polymer
L21 5µm 150x4.6
N = 1,163
2
3
1 4

2 4 6 8 10

Silica
L1 5µm 150x4.6 N = 18,992 Isocratic:80:20 ACN/Water
3 Flow: 1mL/min
2
• Uracil
• Acetophenone
• Toluene
1 • Acenaphthalene

2 4 6 8 10
48
© 2018 USP
Hybrid Particles

 Mixture of Silica and Polymer


– Mixed throughout
– Surface coated

 Higher mechanical stability


 Efficiency more similar to Silica

 pH stability 1-12
 Easier to modify, more stationary
phases available

49
© 2018 USP

Efficiency Comparisons

2 Silica L1 5µm 150 x 4.6 mm


N = 84,573 p/m

1 Mobile phase: 50:50 Acetonitrile/


3 4 20 mM NaH2PO4, pH 2.5
5
Flow rate: 1.0 mL/min
Detection: 230 nm
Components: 1. Ethylparaben
2. Naproxen
4 6 8 10 12 14
3. Diflunisal
Time (min)
4. Indomethacin
2 5. Ibuprofen
Hybrid L1 5µm 150 x 4.6 mm
N = 80,480 p/m
1
3
4 5

2 4 6 8 10 12 14
Time (min) 50
© 2018 USP
Effect of Particle Size at 30°C

28 1 .9 u 3 0 C
2 .5 u 3 0 C
26
3u30C
24 5u30C
22

20
Plate Height µm

18

16

14 5µm
12

10
3µm
8
2.5µm
6
1.9µm
4
0 2 4 6
L in e a r V e lo c ity ( m m /s e c )
51
© 2018 USP

General Theory

L
 Performance Increase N =
hd p

N  α − 1  k 
 Resolution Increase R =   
4  α  k + 1 

φη L µ
 Backpressure Increase ∆P = 2
µ = linear velocity d p
L = column length
dp = particle diameter 52
© 2018 USP
Fully Porous Particles

Standard Pressures =
Conventional HPLC

5 µm 3 µm
10 µm
Operating Pressure up to 400 bar (6,000 psi)

Ultra-High Pressure =
UHPLC
sub-2 µm
Pressure 400-1000 bar (15,000 psi)

53
© 2018 USP

Monolithic Columns

Não é possív el exibir esta imagem.

54
© 2018 USP
Core Shell Columns

 Non-porous silica core with thin porous shell


 Thin shell reduces the diffusion path length
 Result: ultra-fast kinetics of diffusion
 Efficiencies similar to sub 2 µm fully porous particles
 Back pressure similar to 3 µm

55
© 2018 USP

Columns

What brand of column was used for a particular test?


− If you have access to USP–NF on-line scroll to the end of the monograph containing the test of
interest and click on the link under the heading Chromatographic Column
• You can also find Chromatographic Columns on-line database on the USP website at:
http://www.usp.org/resources/chromatographic-columns

56
© 2018 USP
USP–NF Online

57
© 2018 USP

www.uspchromcolumns.com

58
© 2018 USP
www.uspchromcolumns.com

59
© 2018 USP

Briefing in PF

60
© 2018 USP
Columns

Column Equivalency Compendial Tool:


http://apps.usp.org/app/USPNF/columns.html

An interactive tool that allows users to find possible alternative HPLC columns to
the column used to develop and validate a chromatographic procedure.

61
© 2018 USP

62
© 2018 USP
63
© 2018 USP

Column Characterization

 Two approaches cited by USP:


– USP database: NIST SRM 870 mix
– PQRI/Column equivalence database

 Other theories exist – None is 100%

64
© 2018 USP
SRM 870

 Void Marker:
– Unretained volume required to calculate retention factor

 Methylene Selectivity:
– Differentiation based on differences in hydrophobicity
 Efficiency (N):
– Toluene or Ethylbenzene

 Peak Asymmetry:
– Chelating Agent: indicator of metals in silica
 Silanol Activity:
– Active base that interacts with free silanols
– Peak shape is a measure of column deactivation

65
© 2018 USP

SRM 870 Conditions

Columns: L1 10 µm 250 x 4.6 mm Type B


L1 10 µm 250 x 4.6 mm Type A
Mobile phase: Isocratic with 80/20 MeOH / 20 mM KH2PO4 buffer at pH
7.0 for 25 min
Flow rate: 1.0 mL/min
Injection Vol.: 2.0 µL
Detection: 254 nm

66
© 2018 USP
SRM 870: Type B

Peak Name
1 Uracil
mAU

2 Toluene
Efficiency 3 Ethyl Benzene
200

Void + 4 Quinizarin
Marker Metal Chelator
Methylene 5 Amitriptyline
150
1 Selectivity
4
100
2 3 Silanol Activity

50
5

0 5 10 15 20

67
© 2018 USP

Metal Chelation

Si O H
O O

Si O O

3+ HO
Al Chelation

HO
Si O
CO O H
O
HO O C
Si
O
Si O H

68
© 2018 USP
SRM 870: Type A

Quinizarin
Peak Name

1 Uracil
200 2 Toluene
3 Ethyl Benzene
150
High Metal Content 4 Quinizarin
1 Silica 5 Amitriptyline
100

2 3
50

5
4
0

0 5 10 15 20

69
© 2018 USP

PQRI Database

 Proposed by Synder/Dolan
 Columns are evaluated for:
– Hydrophobicity (H)
– Steric interaction (S)
– Hydrogen-bond acidity (A)
– Hydrogen-bond basicity (B)
– Ion-exchange capacity (C)

 Two pH choices - Low 2.8 or High 7.0


 Similarity given by an F rating:
– F < 3 are likely to give equivalent results

70
© 2018 USP
PQRI Database Cont.

71
© 2018 USP

Column Maintenance

 Follow manufacturer’s recommendations


 In general, for reversed phase columns, routinely flush with strong mobile phase
component. Flush buffers with water/organic mixture in the same ratio as the
mobile phase, and store columns in water/organic mixture such as methanol or
acetonitrile and water (1:1)
 Use a log book
 Use the reference chromatogram and standard conditions to check column
performance
 Segregate columns by use (mobile phase, product, test, etc.)
 Use an inline filter
72
© 2018 USP
Column Maintenance: Use of Guard Column

“In LC procedures, a guard column may be used with the following


requirements, unless otherwise is indicated in the individual monograph:

the length of the guard column must be NMT 15% of the length of the analytical column,
(a)
and

(b)the packing material should be the same as the analytical column (e.g., silica) and contain
the same bonded phase (e.g., C18).

In any case, all system suitability requirements specified in the official


procedure must be met with the guard column installed”
73
© 2018 USP

Standard and Sample Preparation

 The dilution scheme can be changed, but the final concentration of the analyte
can not change more than 10% -- see General Notices
 Glassware sizes, weights, volumes, concentrations can be changed
 Errors associated to dilutions and weighing (see USP <31> and <41>)

74
© 2018 USP
Detector

 Spectrophotometric
– Variable
• Single wavelength
• Dual wavelength
– Multiwavelength

• Diode array

 Refractive Index
 Fluorometric
 Electrochemical
 MS
75
© 2018 USP

Quantitation

 During quantitation, disregard peaks caused by solvents and reagents or arising


from the mobile phase or the sample matrix.
 In the linear range, peak areas and peak heights are usually proportional to the
quantity of compound eluting. The peak areas and peak heights are commonly
measured by electronic integrators but may be determined by more classical
approaches. Peak areas are generally used but may be less accurate if peak
interference occurs.
 In tests for impurities for both the External Standard Method, when a dilution of
the sample solution is used for comparison, and the Normalization Procedure,
any correction factors indicated in the monograph are applied (e.g., when the
relative response factor is outside the range 0.8–1.2).

76
© 2018 USP
Quantitation

 When the impurity test prescribes the total of impurities or there is a quantitative
determination of an impurity, choice of an appropriate threshold setting and
appropriate conditions for the integration of the peak areas is important. In such
tests the limit at or below which a peak is disregarded is generally 0.05%. Thus,
the threshold setting of the data collection system corresponds to at least half of
this limit. Integrate the peak area of any impurity that is not completely
separated from the principal peak, preferably by valley-to-valley extrapolation
(tangential skim).

77
© 2018 USP

Methods of Integration

A C

B D

Bicking, M. LCGC NORTH AMERICA VOLUME 24 NUMBER 4 APRIL 2006


78
© 2018 USP
Periodic Instrument Check for HPLC

 Pump (solvent delivery system)


– Flow rate accuracy
– Gradient composition accuracy

 Injector
– Volume precision
– Carry-over
– Thermostating accuracy and precision (Auto sampler)

79
© 2018 USP

Periodic Instrument Check for HPLC (cont.)

 Detector
– Linearity
– Wavelength accuracy
– Noise and drift
– Sensitivity (Florescence)
– Signal stability (Electrochemical)

 Column compartment (Oven or cooling device)


– Temperature accuracy

80
© 2018 USP
Compendial HPLC Practices
System Suitability
System Suitability

 …used to verify that the chromatographic system is adequate for the intended
analysis.

 The Chromatographic system includes:


– Equipment
– Electronics
– Analytical Operations
– Samples

83
© 2018 USP

Parameters

 Resolution
 Efficiency
 Precision
 Tailing
 Retention Factor (Capacity Factor)
 Signal to Noise ratio
 Peak to valley ratio

84
© 2018 USP
Resolution

Resolution, Rs, ensure that closely eluting compounds are


resolved from each other

2 ( t Rb − t Ra ) 1 . 18 ( t Rb − t Ra )
R = R =
Wa + Wb W 0 .5 a + W 0 .5 b

85
© 2018 USP

Resolution

 Function of column efficiency (N), the separation factor, and the capacity factor,
k
 Measure of the resolving power of the system
 Generally, not less than 2.0
 Most closely eluting pair

86
© 2018 USP
Resolution

R = 1.0 R = 1.5 R = 2.0


FDA (CDER), Reviewer Guidance: Validation of Chromatographic Methods, Nov. 1994

87
© 2018 USP

Separation as a Function of Rs

88
© 2018 USP
Column Efficiency

2
 t 
2
 
N = 16  R   t
N = 5 . 54  R

W  Wh 
 2 

89
© 2018 USP

Column Efficiency

Method Peak width Factor

Inflection point 60.3% of peak height 4.000

Half-height 50% of peak height 5.545

4 Sigma method 13.4 of peak height 16.00

5 sigma method 4.4% of peak height 24.00

Intersection of tangents with


Tangent method 16.00
baseline

90
© 2018 USP
Column Efficiency

 Only one peak of interest


 Measure of peak sharpness
 Detection of trace components
 Generally, not less than 2000 (HPLC)
 Isocratic/isothermal system

91
© 2018 USP

Repeatability

 USP: Data from five replicate injections of the analyte are used to calculate the
relative standard deviation if the requirement is 2.0% or less; data from six
replicate injections are used if the relative standard deviation requirement is
more than 2.0%

Number of individuals injections


3 4 5 6
Upper limit Maximum permitted relative standard deviation
2.0 0.41 0.59 0.73 0.85
2.5 0.52 0.74 0.92 1.06
3.0 0.62 0.89 1.10 1.27

92
© 2018 USP
Asymmetry Factor (Tailing)

(a + b) b'
T = A =
2a a' T A
1.0 1.0

1.2 1.3

1.4 1.6

1.6 1.9

a’ 1.8 2.2
b’ 10 % of Peak Height

a b 5 % of Peak Height

93
© 2018 USP

Retention Factor (or Capacity Factor, k)

 Measure of where the peak of interest is located with respect to the void volume,
i.e., elution time of the non-retained components
 Generally, not less than 2

 k = (tR - tM)/ tM
 tR: retention time of the analyte
 tM: retention time of a non-retained component

94
© 2018 USP
Signal to Noise Ratio (S/N)

S/N = 2H/h

95
© 2018 USP

Peak-to-Valley Ratio

p /v = H p H v

96
© 2018 USP
System Suitability USP <621>

 Unless otherwise directed in the monograph, system suitability parameters are


determined from the analyte peak.
 Measured values of Rr or RF or tR for the sample substance do not deviate from
the values obtained for the reference compound and mixture by more than the
statistically determined reliability estimates from replicate assays of the
reference compound.
 Relative retention times may be provided in monographs for informational
purposes only to aid in peak identification. There are no acceptance criteria
applied to relative retention times.

97
© 2018 USP

System Suitability USP <621>

 Make injections of the appropriate preparation(s) as required in order to


demonstrate adequate system suitability (as described in the Chromatographic
system section of the method in a monograph) throughout the run.
 Whenever there is a significant change in the chromatographic system
(equipment, mobile phase component, or other components) or in a critical
reagent, system suitability is to be reestablished.
 No sample analysis is acceptable unless the suitability of the system has been
demonstrated

98
© 2018 USP
Compendial HPLC Practices
Adjustments to the Chromatographic System
Adjustments to the Chromatographic System

 USP <621> provides allowable adjustments to:


– Mobile phase composition
– Mobile phase pH
– Buffer concentration
– Column temperature
– Injection volume
– UV-VIS detector wavelength
– Particle size
– Column length
– Column inner diameter
– Flow rate
101
© 2018 USP

Adjustments: Isocratic vs. Gradient

“Adjustments to the composition of the mobile phase in gradient elution may


cause changes in selectivity and are not recommended. If adjustments are
necessary, change in column packing (maintaining the same chemistry), the
duration of an initial isocratic hold (when prescribed), and/or dwell volume
adjustments are allowed. Additional allowances for gradient adjustment are
noted in the text below.”

102
© 2018 USP
Adjustments: Isocratic vs. Gradient

Adjustment Isocratic Gradient

pH of Mobile Phase Allowed Allowed


Concentration of Salts in Buffer Allowed Allowed
Allowed Not allowed
Ratio of Components in Mobile Phase

Column dimensions (length, diameter) Allowed Not allowed


Particle size Allowed Not allowed
Flow rate Allowed Not allowed
Injection volume Allowed Allowed
Column temperature Allowed Allowed

103
© 2018 USP

Adjustments to the Chromatographic System

 Must show improvement in chromatography

 Cannot be made where there is column failure or system malfunction

 Adjustment may require verification

 Consider multiple adjustments carefully

104
© 2018 USP
Adjustments to the Chromatographic System

 Adjustments made to the chromatographic system in order to meet system


suitability requirements cannot be incorporated into the method.

 Changes made to the chromatographic system because of the use of columns


with different particle sizes can be incorporated into the method. (example,
changing from HPLC to UHPLC).

105
© 2018 USP

Mobile Phase Composition

 Allowable Adjustment:
– Minor component (≤50%) ± 30% relative
– Adjustment cannot exceed ± 10% absolute
– Final concentration cannot be reduced to zero
– Changes to gradient methods are not recommended

 Effect:
– Solvent strength can increase or decrease retention
– Gradient must be scaled if column dimensions change

106
© 2018 USP
Mobile Phase Composition

 Example (binary)
 Specified ratio of 50:50
– 30% of 50 is 15% absolute (too much),
– Maximum allowable change is 10%
– Allowable Adjustments: 40:60 to 60:40

 Specified ratio of 2:98


– 30% of 2 is 0.6% absolute
– Well within Max 10%
– Allowable Adjustments: 1.4:98.6 to 2.6:97.4
107
© 2018 USP

Mobile Phase Composition

 Example: Ternary Mixtures


 Specified ratio 60:35:5 (A:B:C)
– Component B:
• 30% of 35 is 10.5% absolute, exceeds max 10%
• Adjusted only within the range of 25% to 45% (± 10%)
– Component C:
• 30% of 5 is 1.5% absolute
– Component A:
• Sufficient quantity used to give a total of 100%

108
© 2018 USP
Mobile Phase pH

 Allowable Adjustment:
– pH can be changed ± 0.2 units

 Effect
– pH scale is logarithmic, 1 unit =10X increase
– Low pH can protonate acidic compounds (silanols)
– High pH can neutralize basic compounds
– Ensure pH keeps the drug in one state
• Avoid pH = pKa

109
© 2018 USP

Performance at Different pH

110
© 2018 USP
Buffer Concentration

 Allowable Adjustment:
– Buffer concentration ± 10%
– Provided permitted pH variation is also met

 Effect
– If buffer concentration is insufficient, pH changes may occur depending on injection
– Concentration is most critical at pH extremes of that buffer

111
© 2018 USP

Column Temperature

 Allowable Adjustment:
– Column temperature ± 10°C

 Effect
– Temperature can cause shifts in retention time
– Ambient Temp. is relative, it should be controlled
– Higher Temp. decreases mobile phase viscosity
• Improves column efficiency
• Lower column back pressure
• Decrease analysis time

– These changes may affect resolution

112
© 2018 USP
Changing Temperature

Method: Nitroaromatics

Mobile phase: 55:45 MeOH/Water hold 2 min


to 75:25 in 22 min

Flow rate: 1.0 mL/min

Columns used: C18, 250 x 4.6 mm, 4 µm

Temperature: 25 °, 30 °, 35 °

113
© 2018 USP

Column Temperature

3
Temperature: 25 °C

13
7 8
1 5
2 14

4 6 9 10 11 12

10 12 14 16 18

114
© 2018 USP
Column Temperature

Elution Order
Co-Elution
Change Temperature: 30 °C

5,7
2,3
Decreased
1 8 Retention
6 13

14
4 9 10 11
12

8 10 12 14

115
© 2018 USP

Column Temperature

Co-Elution

Elution Order
5,6,7 Temperature: 35 °C
Change

3 Co-Elution
Decreased
Retention
13
8 11,12
1 2
14

4 9 10

18 10 12 14

116
© 2018 USP
Injection Volume

 Allowable Adjustment:
– The injection volume can be adjusted as far as it is consistent with accepted precision, linearity, and
detection limits.
– Note that excessive injection volume can lead to unacceptable band broadening, causing a reduction
in N and resolution.
– Applies to both gradient and isocratic separations.

117
© 2018 USP

UV/VIS Detector Wavelength

 Allowable Adjustment:
– No deviation permitted
– If detector is suspected
• Verify detector wavelength ±3 nm
• Use manufacture or other validated protocol

118
© 2018 USP
Particle Size

 For isocratic separations, the particle size and/or the length of the column may
be modified provided that the ratio of the column length (L ) to the particle size
(dp ) remains constant or into the range between - 25% to +50% of the
prescribed L /dp ratio.

 Alternatively (as for the application of particle-size adjustment to superficially


porous particles), other combinations of L and dp can be used provided that the
number of theoretical plates (N ) is within -25% to +50%, relative to the
prescribed column.

119
© 2018 USP

Particle Size

Flow Rate: When the particle size is changed, the flow rate may require
adjustment:
F2 = F1 (dc22 dp1) / (dc12 dp2)

Where F1 and F2 are the flow rates for the original and modified conditions,
respectively; dc1 and dc2 are the respective column diameters, and dp1 and dp2 are
the particle sizes.

120
© 2018 USP
Particle Size

dc 22 × dp 1
F 2 = F1
dc 12 × dp 2
Relative Values
L, mm dc, mm dp, µm L/dp F N Pressure Time
250 4.6 10 25,000 0.5 0.8 0.2 3.3
150 4.6 5 30,000 1.0 1.0 1.0 1.0
150 2.1 5 30,000 0.2 1.0 1.0 1.0
100 4.6 3.5 28,600 1.4 1.0 1.9 0.5
100 2.1 3.5 28,600 0.3 1.0 1.9 0.5
75 4.6 2.5 30,000 2.0 1.0 4.0 0.3
75 2.1 2.5 30,000 0.4 1.0 4.0 0.3
50 4.6 1.7 29,400 2.9 1.0 8.5 0.1

Transfer of HPLC Procedures to Suitable Columns of Reduced Dimensions and Particle Sizes. Pharm. Forum 35(6), www.usppf.com
121
© 2018 USP

Particle Size

122
© 2018 USP
Column ID

 Allowable Adjustment:
– Can be adjusted provided that linear velocity is kept constant

F2 = F1 (dc22 dp1) / (dc12 dp2)


 Effect
– Solvent savings
– Reduced band broadening (sharper peaks)
– Increased resolution (sharper peaks)

123
© 2018 USP

Column ID

Method: Atenolol Impurity

Mobile phase: Isocratic: 100% of 2.0g of Sodium Octanesulphonate + 0.8g of Tetrabutyl


Ammonium Hydrogen Sulphate in 2L of 360ml Methanol + 40ml of
THF+1600ml of 3.4g of potassium Dihydrogen Phosphate in 2L of water
(pH adjusted to 3.0 using Phosphoric acid)

Flow rate: 0.8 & 0.36 mL/min (scaled for linear velocity)

Columns used: L1 5µm 150 x 4.6* mm


L1 5µm 150 x 3.0* mm

System suitability: Rs = Impurities I & J > 1.4


124
© 2018 USP
Column ID
mAU

B
8 Rs =3.81
6
4.6 mm
J Solvent Usage: 28 mL
4

2 I F
G
0

5 10 15 20 25 30 35

Rs =3.75
mAU
B
J
8

2 X Sensitivity 3.0 mm
6 Solvent Usage: 12.6 mL
4
I
2
F
G
0

5 10 15 20 25 30 35
125
© 2018 USP

Changing Column ID

Summary: Decrease column ID by 35%:

1. Retention time: About the same

2. Solvent Usage: Decreased by 57%

3. Sensitivity: ~ 2X Increase

4. System suitability: Retained Rs > 1.4 from excipients

126
© 2018 USP
Flow Rate

 Allowable Adjustment:
– Flow rate ± 50%
– If changing particle size and/or ID scale to maintain linear flow velocity
– Scaled flow can then be adjusted ± 50%

 Benefit
– Increasing flow is the easiest way to reduce run time
– Isocractic: 2 X Flow = ½ Run Time
– Resolution decreases, but not as dramatically
– Faster linear velocity is required with smaller particles
127
© 2018 USP

Changing Flow Rate

Method: Estradiol

Mobile phase: Water / Acetonitrile (45:55)

Flow rate: 1.0 and 1.5 mL/min

Columns used: L1 5µm 250 x 4.6 mm

System suitability: Rs; Estradiol and Estrone > 2.0

128
© 2018 USP
Changing Flow Rate

1
mAU
1.0 mL/min
350
1. Ethyl Paraben (IS)
300
2. Estradiol
250
3. Estrone 2
200 Rs = 7.11
150 tR = 8.15 min
100

50 3
0

1 2 3 4 5 6 7 8 9 min

1
1.5 mL/min

2 Rs = 6.15
tR = 5.56 min
3

129
© 2018 USP

Changing Flow Rate

Summary: Increasing flow rate 50%

1. Retention time: Reduced from 8.15 to 5.56 min


32% reduction

2. Resolution: Reduced from 7.11 to 6.15


13.5% reduction

3. System suitability: Maintained Rs > 2.0

130
© 2018 USP
Questions (True/False)

 A. The acceptable variation for relative retention time is about +/-10%.

 B. p/v ratio is the preferred term for calculation of peak resolution.

 C. The repeatability guideline in USP <621> needs to be followed in all cases


because it is a mandatory general chapter.

132
© 2018 USP
Questions (True/False)

 D. The repeatability criteria in USP <621> should are used for the drug product
only if this criteria is not in the monograph.

 E. Adjustments to the chromatographic system is only allowed for isocratic


procedures.

 F. Adjustments to the chromatography system is allowed to provide flexibility for


the column failure.

133
© 2018 USP

Compendial HPLC Practices


Harmonization
Pharmacopoeial Discussion Group (PDG)

 USP has participated since PDG’s inception in 1989


 Informal body of representatives from
– European Pharmacopoeia (EDQM)
– Japanese Pharmacopoeia (MHLW)
– United States Pharmacopeia (non-governmental)
– WHO (observer since 2001)
 Linked to ICH (until 2011)
 Focused on harmonizing general chapters and excipient monographs
 Steady but limited progress (Retrospective Harmonization)
– 29 out of 36 General Chapters
– 46 out of 62 Excipients

135
© 2018 USP

PDG Stages of Harmonization

136
© 2018 USP
Liquid Chromatography: Isocratic Elution

Column parameters and flow rate

 Stationary phase: no change of the physico-chemical characteristics of the


stationary phase permitted,
 Column dimensions:
– The particle size and/or length of the column may be modified provided that the ratio of the
column length (L) to the particle size (dp) remains constant or in the range between -25 per
cent to +50 per cent of the prescribed L/dp ratio.. Further adjustments in method conditions
(mobile phase, temperature, pH, etc.) may be required, within the permitted ranges described
under System Suitability and Adjustment of chromatographic conditions in this chapter.

137
© 2018 USP

Liquid Chromatography: Isocratic Elution

 When the particle size is changed, the flow rate may require
adjustment, because smaller-particle columns will require higher
linear velocities for the same performance (as measured by reduced
plate height). Flow rate changes for both the change in column
diameter and particle size can be made by:

F2 = F1 (dc22 dp1) / (dc12 dp2)

138
© 2018 USP
Liquid Chromatography: Isocratic Elution

Injection volume
When the column dimensions are changed, injection volume adjustment may be guided by,
Vinj,2 = Vinj,1 (L2 dc,22) / (L1 dc,12)
Vinj,1 = original injection volume
Vinj,2 = new injection volume
L1 = original column length
L2 = new column length
dc,1 = original column internal diameter
dc,2 = new column internal diameter: ,
Independent of changing column dimensions, Injection volume may be varied provided System Suitability criteria
remain within their established acceptability limits. When Injection volume is decreased, special attention should be
given to (limit of) detection and repeatability of the peak(s) to be determined. An increase is permitted provided in
particular linearity and separation of the peak(s) to be determined remain satisfactory
139
© 2018 USP

Liquid Chromatography: Gradient Elution

 A change in column dimensions, and thus in column volume, impacts the gradient volume which
controls selectivity. Gradients are adjusted to the column volume by changing the gradient volume in
proportion to the column volume. This applies to every gradient segment volume. Because the
gradient volume is the gradient time, tG, multiplied by the flow rate, F, the gradient time for each
gradient segment needs to be adjusted to maintain a constant ratio of the gradient volume to the
column volume (expressed as L x dc2). Thus, the new gradient time, tG2 can be calculated from the
original gradient time, tG1, the flow rate(s), and the column dimensions as follows:
tG2 = tG1 x (F1 / F2) [(L2 x dc22) / (L1 x dc12)]
 Thus, the change in conditions for gradient elution requires three steps:
– (1) Adjust the column length and particle size according to L/dp,
– (2) Adjust the flow rate for changes in particle size and column diameter, and
– (3) Adjust the gradient time of each segment for changes in column length, diameter and flow rate. The
example below illustrates this process.

140
© 2018 USP
Liquid Chromatography: Gradient Elution

Variable Original Adjusted Conditions Comment


Conditions
Column length (L) in mm 150 100 user choice
Column diameter (dc) in mm 4.6 2.1 user choice
Particle size (dp) in µm 5 3 user choice
L/dp 30.0 33.3 (1)
Flow rate in mL/min 2.0 0.7 (2)
Gradient adjustment factor 0.4 (3)
Gradient conditions
%B Time (min) Time (min)
30 0 0
30 3 (3x0.4)=1.2
70 13 [1.2+(10x0.4)]=5.2
30 16 [5.2+(3x0.4)]=6.4

(1) 11% increase within allowed L/dp change of -25% to +50%


(2) calculated using F2= F1 [(dc22 x dp1) / (dc12 x dp2)]
(3) calculated using tG2 = tG1 x (F1 / F2) [(L2 x dc22) / (L1 x dc12)]
141
© 2018 USP

Liquid Chromatography: Gradient Elution

Injection volume

When the column dimensions are changed, injection volume adjustment may be
guided by,

Vinj,2 = Vinj,1 (L2 dc,22) / (L1 dc,12)

Independent of changing column dimensions, injection volume may be varied


provided System Suitability criteria remain within their established acceptability
limits. When Injection volume is decreased, special attention should be given to
(limit of) detection and repeatability of the peak(s) to be determined. An
increase is permitted provided in particular linearity and separation of the peak(s)
to be determined remain satisfactory
142
© 2018 USP
Harmonization – Next Steps

 New proposal published in PF 43(5)

 Comments are being collected and summarized

 Comments will be sent to Eur. Pharm. (coordinator)

143
© 2018 USP

Sources of information

www.usppf.com

www.chromatographyonline.com

www.uspchromcolumns.com

144
© 2018 USP
Click to edit
Validation
master title style

Definitions of Validation: USP

 USP <1225>:
– “Validation of an analytical procedure is the process by which it is established, by laboratory
studies, that the performance characteristics of the procedure meet the requirements for the
intended analytical applications.”

146
© 2018 USP
Validation Characteristics

 Accuracy
 Precision
 Specificity
 Detection Limit
 Quantitation Limit
 Linearity
 Range
Not part of the formal validation study:
 Robustness
 Sample solution stability
147
© 2018 USP

USP <1225> Validation of Compendial Procedures

Performance Category I Category II Category III Category IV


Characteristic
Quant Limit
Test

Accuracy Yes Yes * * No


Precision Yes Yes No Yes No

Specificity Yes Yes Yes * Yes

Detection Limit No No Yes * No

Quantitation Limit No Yes No * No

Linearity Yes Yes No * No

Range Yes Yes * * No


* May be required, depending on the nature of the specific test

148
© 2018 USP
ICH Q2 (R1)

Performance Assay Impurities Identification


Characteristic
Quant Limit Test

Accuracy Yes Yes No No

Precision
Repeatability Yes Yes No No
Interm. Precision Yes (1) Yes (1) No No

Specificity (2) Yes Yes Yes Yes

LOD No No (3) Yes No

LOQ No Yes No No

Linearity Yes Yes No No

Range Yes Yes No No


(1) in cases where reproducibility has been performed, intermediate precision is not needed
(2) lack of specificity of one analytical procedure could be compensated by other supporting procedure(s)
(3) may be needed in some cases
149
© 2018 USP

Specificity

Definition – The method’s ability to specifically measure the analyte in the


presence of the components in the analytical matrix

 Impurities, degradation products and/or excipients are available.

 When impurities, degradation products or excipients are not available:


– Compare the results with another well-characterised analytical method
– Perform an artificial degradation under drastic conditions (light, heat, acids y bases, etc.)

150
© 2018 USP
Specificity

 Required for the validation of


– Identification Tests(category IV)
– Impurity Tests (category II)
– Assay (category I)
– Performance Tests (category III)

 In the pharmacopeial analysis, the lack of specificity of a method can be supplemented


with another to obtain the desired total level of selectivity

 IUPAC, AOAC prefer the term selectivity, reserving the term specificity for fully selective
procedures
151
© 2018 USP

Specificity

 Peak purity

 Peak shape analysis

 Diode array detection

 HPLC-MS

152
© 2018 USP
Forced Degradations

 In solution and/or in solid phase

 The conditions are more severe than in an accelerated stability study

 One Lot

 Mechanisms of thermolysis, hydrolysis, oxidation and photolysis in the drug and


product

 PDA or MS Detectors are necessary for validation, not for QC


153
© 2018 USP

Forced Degradations

Guidelines for submitting samples and analytical data for methods validation,
FDA, 1987.
 Drug substances:
– Heat (50°C)
– Light
– Hydrolysis: acid (HCl 0.1 N) / base (NaOH 0.1 N)
– Oxidation (H2O2 3%)
 Drug Products
– Heat
– Light
– Humidity (85%)
 Other conditions may be necessary
 Exposure time, temperature and concentration can be adjusted to obtain an
appropriate degradation of the sample (10% to 15%)
154
© 2018 USP
Linearity

Definition – The ability of a method to produce results that are directly proportional
to the concentration of the sample at a given interval

 5 levels of concentration. Different dilutions of an stock solution or separate


weights of the component

 3 samples per level of concentration

155
© 2018 USP

Linearity

Calibration model Requirements


Single-point calibration • Linear response function
• Negligible systematic error
• Homosedasticity
Multiple-point calibration (linear, unweighted) • Linear response function
• Homosedasticity
Multiple point calibration (linear, weighted) • Linear response function

Multiple point calibration (non-linear) • Continuous response function


Area normalization • For main peak
‒ Linear response function
‒ Negligible systematic error
‒ Homosedasticity
• For impurities
‒ Linear response function
‒ Negligible systematic error
156
© 2018 USP
Linearity

y = a + bx
 [( x − x )( y − y ]
i
i i
b=
 (x − x)i
i
2

a = y − bx
157
© 2018 USP

Coefficient of Correlation

 [( x − x )( y
i
i i − y )]
r=
 2  
 
 i
( xi − x )  
 i
( y i − y ) 

158
© 2018 USP
Confidence Intervals

(y i
i − yˆ i ) 2
Sy x =
n−2

Sy x
b ± t(n-2) Sb
Sb =
 i
( xi − x ) 2 a ± t(n-2) Sa

 i
x i2
Sa = Sy
n  ( xi − x ) 2
x

159
© 2018 USP

Residue Analysis

20
15
10
Residuals

5
0
-5
-10
-15
-20
0 5 10 15 20 25 30 35
Cc (mg/ml)

 Check that the graph has a random pattern.


 A trend indicates a failure in the chosen model.
 An increase or decrease in residue dispersion indicates that the data is not homoscedastic.

160
© 2018 USP
Residue Analysis

Normal

Pattern Increasing trend

161
© 2018 USP

Residue Analysis

 Weighted Linear Regression

– The homoscedasticity is a prerequisite for the common lineal regression

– In LC-UV it is generally assumed that this occurs within a range of one order of magnitude

– In greater concentration ranges it would probably violate this assumption

– Ifrequired to quantify a large range of concentrations (more than two orders of magnitude) the
lack of homoscedasticity should be compensated by a weighted linear regression
162
© 2018 USP
Impurity Analysis – Various Types

Area normalization
 Individual impurity is calculated as follows:

(Areaimp/Areatotal) 100

 Useful in the initial stages of development. Does not require a reference standard
 Requires linearity over a wide range
 If the concentration is reduce to maintain linearity, impurity can not be detected
 Assume the response factors are similar
 Accuracy evaluation is not required
 Example: Dousa M., Gibala P, Pekarek T – New approach of validation using internal
normalization technique for quantification of related substances in raw materials, intermediates
and pharmaceutical substances by HPLC. J. Pharm. Biom. Anal. 114(2015): 133 – 138.
163
© 2018 USP

Impurity Analysis – Various Types

“High-Low”

 A concentrated sample solution compared against a diluted sample solution


 Removes limitation of the previous method with respect to linearity
 The peaks are compared in a limited concentration range

164
© 2018 USP
Impurity Analysis – Various Types

External Standard

 The concentration is determined from a calibration curve


 Uses a reference standard or assumes that the response factors are equal
 The single-point calibration is appropriate if the y-intercept is not significant
 Uses a small linear range
 Improve sensitivity

165
© 2018 USP

Accuracy

Definition - The difference between results obtained by the method and the actual
value.

 It is calculated by determining the % recovery of the method


Cobs × 100
R=
Cad

 Three replicates at three levels


– 80% -120% for the principal component
– 50% - 150% for impurities

166
© 2018 USP
Accuracy

 Drug substance
– Analysis of reference material

 Drug product
– Analysis of synthetic mixtures

 Impurities
– Analysis of samples spiked with known amounts of impurities

 Compare the result with a second well-characterized method


167
© 2018 USP

Recovery Test

40
y = 0.9989x + 0.3272
R² = 0.9974 Amount added (mg) Amound detected (mg) Recovered
24.35 24.70 101.4%
35
25.15 25.41 101.0%
25.15 25.17 100.1%
observed

30.04 30.79 102.5%


30
29.74 30.18 101.5%
30.14 30.38 100.8%
25 35.33 35.70 101.0%
34.63 34.56 99.8%
Ecuacion de la rect a
36.73 37.01 100.8%
20 rado = a + ( b x Añadido)
Encont Mean: 101.0%
20 25 b= 30 35
0.9989
added SD: 0.7945
a= 0.3272

168
© 2018 USP
Recovery Test – USP <1225>

Assessment of accuracy can be accomplished in a variety of ways,


including evaluating the recovery of the analyte (percent recovery) across the
range of the assay, or evaluating the linearity of the relationship between
estimated and actual concentrations. The statistically preferred criterion is that
the confidence interval for the slope be contained in an interval around 1.0, or
alternatively, that the slope be close to 1.0. In either case, the interval or the
definition of closeness should be specified in the validation protocol. The
acceptance criterion will depend on the assay and its variability and on the
product. Setting an acceptance criterion based on the lack of statistical
significance of the test of the null hypothesis that the slope is 1.0 is not an
acceptable approach.

169
© 2018 USP

Precision

Definition - The degree of agreement between results when a procedure is


repeatedly applied to a homogenous sample

 Repeatability, Intermediate Precision, Reproducibility

 Calculation of the SD or %RSD

170
© 2018 USP
Precision

 Repeatability: is precision under the same operating conditions for a short


period of time. It is determined by a minimum of 9 measurements within the
given range of the procedure (3 concentrations/ 3 replications) or a minimum of
6 replications at 100%

 Intermediate Precision: indicates intra-laboratory variations; different days,


different analysts, different equipment

 Reproducibility: indicates inter-laboratory variations

171
© 2018 USP

Range

Definition - The difference between the highest and lowest concentrations of


sample (includes these two values) that can be determined with acceptable
precision, accuracy and linearity.

 Assay of Macro-components 80-120%


 Uniformity of Dosage Units 70-130%
 Dissolution Test ± 20%
 Quantification of Impurities 50%-120%

172
© 2018 USP
Range

Response

Linearity and Accuracy

Precision

Range

Concentration

173
© 2018 USP

Detection/Quantification Limits

 Detection Limit – The minimum concentration of analyte that can be detected


under the established experimental conditions

 Quantification Limit – The lower concentration of analyte that can be


determined with acceptable precision and accuracy

174
© 2018 USP
Detection/Quantification Limits

Q3A(R2): Impurities in New Drug Substances

Maximum Daily Reporting Identification Quantification


Dose Threshold Threshold Threshold
</= 2g/día 0.05% 0.1% o 1.0 mg per 0.15% o 1.0 mg per
day (whichever is day (whichever is
lower) lower)
> 2 g/día 0.03% 0.05% 0.05%

175
© 2018 USP

Detection/Quantification Limits

Q3B(R2): Impurities in New Drug Products


Maximum Daily Dose Threshold
Reporting Threshold
≤1g 0.1%
>1g 0.05%
Identification Threshold
< 1 mg 1.0% o 5 μg TDI, whichever is lower
1 mg - 10 mg 0.5% o 20 μg TDI, whichever is lower
>10 mg - 2 g 0.2% o 2 mg TDI, whichever is lower
>2g 0.10%
Qualification Threshold
< 10 mg 1.0% o 50 μg TDI, whichever is lower
10 mg - 100 mg 0.5% o 200 μg TDI, whichever is lower
>100 mg - 2 g 0.2% o 3 mg TDI, whichever is lower
>2g 0.15%
176
© 2018 USP
Basic Approaches to Determination

ICH Q2 lists the following

 Visual
 Signal-to-Noise ratio
 Blank standard deviation and regression line

177
© 2018 USP

Visual

 I can see it!!


 ICH Q2 – 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 (LOD)
– The minimum level at which the analyte can be quantified with acceptable accuracy and
precision (LOQ)
 Seems more appropriate for LOD
 Subject to analyst interpretation

178
© 2018 USP
Signal-to-Noise Ratio

 Q2 and USP <1225> -- Compare


measured signals from samples
with known low concentrations of
10:1
analyte with those of blank
samples

 Requirements
– S/N > 3 (or 2) for LOD
3:1
– S/N > 10 for LOQ

179
© 2018 USP

Signal-to-Noise Ratio

h: range of the background noise in a


chromatogram obtained after injection
or application of a blank, observed
over a distance equal to 5 times the
width at half-height of the peak in the
chromatogram obtained with the
prescribed reference solution and, if
possible, situated equally around the
place where this peak would be found
S/N = 2H/h

180
© 2018 USP
Detection/Quantification Limits

Based on the standard deviation of the response:

LOD= 3.3 S /b
LOQ= 10 S /b
S = Standard deviation of the response
b= Slope of the calibration curve

181
© 2018 USP

Basis for this Approach

+10
SD

5%
Blank
+3.3 Sigma
5% +10 Sigma +3.3
SD

blank response

LOD LOQ

The 3.3 and 10 SD values are intended to separate the distribution of


responses at LOD and LOQ from the distribution of blank samples

Dividing by the slope, b, converts differences in y (response) axis to differences


in x (concentration) axis
182
© 2018 USP
Detection/Quantification Limits

Options for S:
 SD of blank responses
– Analyze various blanks and calculate the SD
 Residual SD from regression line

 i i
(
i
y − y
ˆ ) 2

Sy x =
n−2

183
© 2018 USP

USP <1225> Recommendation

 Whatever method is used, the detection [quantitation] limit should be


subsequently validated by the analysis of a suitable number of samples known
to be near, or prepared at, the detection [quantitation] limit

184
© 2018 USP
Example

Concentration (μg/ml) Peak Area LOD = 3.3 * 0.00033149/0.2920791


0.0100994 0.003108
0.0252486 0.008015
0.0504972
0.0757458
0.015540
0.022415
= 0.0037
0.1009944 0.029510
0.2524860 0.074235
LOQ = 10 * 0.00033149/0.2920791
Source: Table 3-11 of Ermer & Miller

SUMMARY OUTPUT
= 0.0113
Regression Statistics
Multiple R 0.99993354
R Square 0.99986709
Adjusted R Square 0.99983386
Standard Error 0.00033149
Observations 6
Note: “Standard Error” is Residual SD
ANOVA
df SS MS F Significance F
Regression 1 0.003306581 0.003307 30090.49 6.62516E-09
Residual 4 4.39552E-07 1.1E-07
Total 5 0.003307021

Coefficients Standard Error t Stat P-value Lower 95% Upper 95%


Intercept 0.0003969 0.00019801 2.004458 0.115527 -0.000152862 0.0009467
Concentration (μg/ml) 0.2920791 0.001683782 173.4661 6.63E-09 0.287404158 0.296754

185
© 2018 USP

Robustness

Definition – A measure of a method’s ability to accept small but deliberate


variations in the parameters of the method

 Steps to Consider
– Design and execution of the experiment
– Calculating the effects
– Statistical/graphical analysis of the effects
– Relevant conclusions and eventual modifications of the method

 Changes in: pH, column, mobile phase, etc.


186
© 2018 USP
Robustness

Experimental Design
 One factor at a time

 Factorial Designs
– Full Factorial
– Fractional Factorial
– Plackett-Burman

187
© 2018 USP

Full Factorial Design (2k Runs)

188
© 2018 USP
Fractional Factorial Design – 5 Factors

189
© 2018 USP

Placket - Burman

7 factors (A-G) 2 levels


Exp. Factors Result
A B C D E F G
1 + + + + + + + y1
2 + + - + - - - y2
3 + - + - + - - y3
4 + - - - - + + y4
5 - + + - - + - y5
6 - + - - + - + y6
7 - - + + - - + y7
8 - - - + + + - y8

Eff = ( y+ -  y-)/4
190
© 2018 USP
Example – multi-variate

Table A. Factor levels used in the design

Nominal Lower Upper Variation


Factor Value Level (-1) Level (+1) (%)
Gradient Profile 1 0 2 0
Column Temp. (°C) 40 38 42 5
Buffer Conc. (mM) 40 36 44 10
Buffer pH 5 4.8 5.2 4
Wavelength (nm) 446 441 451 1
Triethylamine (%) 0.23 0.21 0.25 8
Dimethylformamide (%) 10 9.5 10.5 5

Romero, R. et. al. - LC-GC, 20(1): 72 - 80, 2002


191
© 2018 USP

Example – multi-variate

Table B. Experimental design for robustness test


RESPONSE VARIABLES
FACTOR Phenylethylamine Tyramine
Buffer Buffer Wave- TEA Peak Peak tr Unadj.
Run Gradient Temp (°C) Conc. pH length (%) DMF (%) Area* Height* (min) tr (min) R
1 +1 +1 +1 +1 +1 +1 +1 2,365 221.4 18.331 1.1121 1.624
2 +1 +1 -1 -1 +1 -1 -1 2,376 226.0 18.370 1.1108 1.673
3 +1 -1 +1 +1 -1 -1 -1 2,305 226.1 18.671 1.1087 1.745
4 +1 -1 -1 -1 -1 +1 +1 2,330 225.1 18.524 1.1106 1.641
5 -1 +1 +1 -1 -1 +1 -1 2,378 221.8 18.504 1.1101 1.693
6 -1 +1 -1 +1 -1 -1 +1 2,384 224.3 18.561 1.1101 1.736
7 -1 -1 +1 -1 +1 -1 +1 2,448 235.0 18.548 1.1132 1.783
8 -1 -1 -1 +1 +1 +1 -1 2,384 234.7 18.853 1.1071 1.787
9 0 0 0 0 0 0 0 2,357 221.2 18.648 1.1086 1.653
10 0 0 0 0 0 0 0 2,311 218.4 18.639 1.1092 1.689
11 0 0 0 0 0 0 0 2,312 219.9 18.667 1.1088 1.733
* Arbitraty Units

Romero, R. et. al. - LC-GC, 20(1): 72 - 80, 2002


192
© 2018 USP
Definitions for Verification: USP

USP <1225>: “…users of analytical methods described in the USP–NF are not
required to validate accuracy and reliability of these methods, but merely verify
their suitability under actual conditions of use.”

USP <1226>: “Verification consists of assessing selected analytical performance


characteristics, such as those that are described in USP <1225>, to generate
appropriate, relevant data rather than repeating the validation process.”

193
© 2018 USP

Definitions for Verification: FDA

 Guide to Inspections of Pharmaceutical Quality Control Laboratories:

 “Methods appearing in the USP are considered validated and they are
considered validated if part of an approved ANDA”

 “For compendial methods firms must demonstrate that the method works under
the actual conditions of use.”

194
194
© 2018 USP
Concepts

Validation: Challenges the analytical method using a well defined sample

Verification: Challenges the analytical environment using a well defined method


– Analyst
– Instrument
– Reagents
– Matrix

195
© 2018 USP

USP <1226> Summary

 The intent of this chapter is to provide general information to laboratories on the


verification of compendial procedures that are being performed for the first time
to yield acceptable results utilizing the laboratories’ personnel, equipment, and
reagents

 Not intended for retroactive application to already successfully established


laboratory procedures

 Verification consists of assessing selected Analytical Performance


Characteristics, such as those which are described in USP chapter <1225>, to
generate appropriate, relevant data rather than repeating the validation process

196
© 2018 USP
A Risk-based Approach

“Verification requirements should be based on an assessment of the complexity of


both the procedure and the material to which the procedure is applied.”

“The degree and extent of the verification process may depend on the level of
training and experience of the user, on the type of procedure and its associated
equipment or instrumentation, on the specific procedural steps, and on which
article(s) are being tested.”

197
© 2018 USP

Common Practices in the Industry

 System suitability requirements


 Specificity –
– Peak purity

– Matrix

– Accuracy

 Accuracy: Recovery in the specification range


 Precision: Repeatability in the specification range

198
© 2018 USP
Common Practices in the Industry (cont)

 LOD: Verification of detection at 50% of the specification


 LOQ: Verification of quantitation at 50% of the specification
 Usually not needed: Linearity, Range, and Robustness

199
© 2018 USP

Common Practices in the Industry

 Other details to be considered

– Columns vs. cartridges

– Mobile phase preparation

– Gradient and dead volume

– Sample solution preparation

– Sample solution stability

200
© 2018 USP
USP <621> Chromatography – Decision Tree

201
© 2018 USP
(301) 230-6304 | http://education.usp.org | Education@usp.org

You might also like