You are on page 1of 66

General Information

KP IX 1817

General Information

1. Capillary Electrophoresis ...................................................................................................... 1818


2. Determination of Bulk and Tapped Densities ..................................................................... 1824
3. Disinfection and Sterilization Methods ................................................................................ 1825
4. Guideline for Setting Dissolution Specification of Oral Dosage Forms ............................ 1827
5. Guideline to Pharmaceutical Quality Control Using Near Infrared (NIR)
Spectroscopy ........................................................................................................................... 1836
6. Guideline of Limits for Residual Solvents of Pharmaceuticals .......................................... 1850
7. Guideline of Validation of Analytical Procedures for Pharmaceuticals ........................... 1857
8. Isoelectric focusing ................................................................................................................. 1862
9. Particle Size Determination .................................................................................................. 1865
10. Powder Particle Density Determination .............................................................................. 1871
11. Preservatives-Effectiveness Tests ......................................................................................... 1872
12. Specific Surface Area Determination Method ..................................................................... 1874
13. Terminal Sterilization and Sterilization Indicators ............................................................ 1876
1818 General Information

velocity of the solutes, both cations and anions can be


1. Capillary Electrophoresis separated in the same run.
The time (t) taken by the solute to migrate the dis-
Capillary electrophoresis is a physical method of tance (l) from the injection end of the capillary to the
analysis based on the migration, inside a capillary, of detection point (capillary effective length) is given by
charged analytes dissolved in an electrolyte solution, the following expression:
under the influence of a direct-current electric field.
The migration velocity of an analyte under an
l lL
electric field of intensity E, is determined by the elec- t 
trophoretic mobility of the analyte and the electro- ep  eo ( ep  eo )  V
osmotic mobility of the buffer inside the capillary. The
electrophoretic mobility of a solute (µ ep) depends on In general, uncoated fused-silica capillaries above
the characteristics of the solute (electric charge, molec- pH 3 have negative charge due to ionized silanol
ular size and shape) and those of the buffer in which groups in the inner wall. Consequently, the electro-
the migration takes place (type and ionic strength of osmotic flow is from anode to cathode. The electro-
the electrolyte, pH, viscosity and additives). The elec- osmotic flow must remain constant from run to run if
trophoretic velocity (ep) of a solute, assuming a spher- good reproducibility is to be obtained in the migration
ical shape, is given by the following equation: velocity of the solutes. For some applications, it may
be necessary to reduce or suppress the electro-osmotic
 q  V  flow by modifying the inner wall of the capillary or by
ep  ep  E    
 r   L 
changing the concentration, composition and/or pH of
 6
the buffer solution. After the introduction of the sample
into the capillary, each analyte ion of the sample mi-
q: effective charge of the solute, grates within the background electrolyte as an inde-
: viscosity of the electrolyte solution, pendent zone, according to its electrophoretic mobility.
r: Stoke's radius of the solute, Zone dispersion, that is the spreading of each solute
V: applied voltage, band, results from different phenomena. Under ideal
L: total length of the capillary. conditions the sole contribution to the solute-zone
broadening is molecular diffusion of the solute along
When an electric field is applied through the ca- the capillary (longitudinal diffusion). In this ideal case
pillary filled with buffer, a flow of solvent is generated the efficiency of the zone, expressed as the number of
inside the capillary, called electro-osmotic flow. The theoretical plates (N), is given by the following equa-
velocity of the electro-osmotic flow depends on the tion:
electro-osmotic mobility (µ eo) which in turn depends
on the charge density on the capillary internal wall and ( ep  eo )  V  l
the buffer characteristics. The electro-osmotic velocity N
(eo) is given by the following equation: 2 D L

D: molecular diffusion coefficient of the solute in


     V 
eo  eo  E       the buffer.
   L
In practice, other phenomena such as heat dissipa-
ε: dielectric constant of the buffer, tion, sample adsorption onto the capillary wall, mis-
: zeta potential of the capillary surface . matched conductivity between sample and buffer,
length of the injection plug, detector cell size and un-
The velocity of the solute () is given by: levelled buffer reservoirs can also significantly con-
tribute to band dispersion.
  ep  eo Separation between two bands (expressed as the
resolution, RS) can be obtained by modifying the elec-
trophoretic mobility of the analytes, the electro-
The electrophoretic mobility of the analyte and osmotic mobility induced in the capillary and by in-
the electro-osmotic mobility may act in the same direc- creasing the efficiency for the band of each analyte,
tion or in opposite directions, depending on the charge according to the following equation:
of the solute. In normal capillary electrophoresis, ani-
ons will migrate in the opposite direction to the electro-
N  ( epb  epa )
osmotic flow and their velocities will be smaller than RS 
the electro-osmotic velocity. Cations will migrate in 4  ( ep  eo )
the same direction as the electro-osmotic flow and their
velocities will be greater than the electro-osmotic ve- µ epa and µ epb: electrophoretic mobilities of the two
locity. Under conditions in which there is a fast electro- analytes separated,
osmotic velocity with respect to the electrophoretic
KP X 1819

 ep : mean electrophoretic mobility of the two electrophoretic mobility of the solute and the electro-
osmotic flow in the capillary. Coated capillaries can be
analytes (ep  1  (epb  epa ))
2 used to increase the separation capacity of those sub-
stances adsorbing on fused-silica surfaces. Using this
Apparatus mode of capillary electrophoresis, the analysis of both
An apparatus for capillary electrophoresis is com- small (Mr < 2000) and large molecules (2000 < Mr
posed of : <100000) can be accomplished. Due to the high effi-
(1) a high-voltage, controllable direct-current power ciency achieved in free solution capillary electrophore-
supply, sis, separation of molecules having only minute differ-
(2) two buffer reservoirs, held at the same level, con- ences in their charge to mass ratio can be effected. This
taining the prescribed anodic and cathodic solutions , separation mode also allows the separation of chiral
(3) two electrode assemblies (the cathode and the an- compounds by addition of chiral selectors to the sepa-
ode), immersed in the buffer reservoirs and connect- ration buffer.
ed to the power supply,
(4) a separation capillary (usually made of fused-silica) Optimization
which, when used with some specific types of detec- Optimization of the separation is a complex pro-
tors, has an optical viewing window aligned with the cess where several separation parameters can play a
detector. The ends of the capillary are placed in the major role. The main factors to be considered in the
buffer reservoirs. The capillary is filled with the so- development of separations are instrumental and elec-
lution prescribed in the monograph, trolytic solution parameters.
(5) a suitable injection system,
(6) a detector able to monitor the amount of substances Instrumental parameters
of interest passing through a segment of the separa- (1) Voltage A Joule heating plot is useful in optimiz-
tion capillary at a given time. It is usually based on ing the applied voltage and column temperature. Sepa-
absorption spectrophotometry (UV and visible) or ration time is inversely proportional to applied voltage.
fluorometry, but conductimetric, amperometric or However, an increase in the voltage used can cause
mass spectrometric detection can be useful for spe- excessive heat production, giving rise to temperature
cific applications. Indirect detection is an alternative and, as a result thereof, viscosity gradients in the buffer
method used to detect non UV-absorbing and non- inside the capillary. This effect causes band broadening
fluorescent compounds, and decreases resolution.
(7) a thermostatic system able to maintain a constant (2) Polarity Electrode polarity can be normal (anode
temperature inside the capillary is recommended to at the inlet and cathode at the outlet) and the electro-
obtain a good separation reproducibility, osmotic flow will move toward the cathode . If the
(8) a recorder and a suitable integrator or a computer. electrode polarity is reversed, the electro-osmotic flow
The definition of the injection process and its au- is away from the outlet and only charged analytes with
tomation are critical for precise quantitative analysis. electro-osmotic mobilities greater than the electro-
Modes of injection include gravity, pressure or vacuum osmotic flow will pass to the outlet.
injection and electrokinetic injection. The amount of (3) Temperature The main effect of temperature is
each sample component introduced electrokinetically observed on buffer viscosity and electrical conductivity,
depends on its electrophoretic mobility, leading to pos- and therefore on migration velocity. In some cases, an
sible discrimination using this injection mode. increase in capillary temperature can cause a confor-
Use the capillary, the buffer solutions, the precon- mational change in proteins, modifying their migration
ditioning method, the sample solution and the migra- time and the efficiency of the separation.
tion conditions prescribed in the monograph of the (4) Capillary The dimensions of the capillary (length
considered substance. The employed electrolytic solu- and internal diameter) contribute to analysis time, effi-
tion is filtered to remove particles and degassed to ciency of separations and load capacity. Increasing
avoid bubble formation that could interfere with the both effective length and total length can decrease the
detection system or interrupt the electrical contact in electric fields (working at constant voltage) which in-
the capillary during the separation run. A rigorous rins- creases migration time. For a given buffer and electric
ing procedure should be developed for each analytical field, heat dissipation, and hence sample band-
method to achieve reproducible migration times of the broadening, depend on the internal diameter of the ca-
solutes. pillary. The latter also affects the detection limit, de-
pending on the sample volume injected and the detec-
1. Capillary Zone Electrophoresis tion system employed. Since the adsorption of the
In capillary zone electrophoresis, analytes are sample components on the capillary wall limits effi-
separated in a capillary containing only buffer without ciency, methods to avoid these interactions should be
any anti-convective medium. With this technique, sep- considered in the development of a separation method.
aration takes place because the different components of In the specific case of proteins, several strategies have
the sample migrate as discrete bands with different been devised to avoid adsorption on the capillary wall.
velocities. The velocity of each band depends on the Some of these strategies (use of extreme pH and ad-
1820 General Information

sorption of positively charged buffer additives) only


require modification of the buffer composition to pre- 2. Capillary Gel Electrophoresis
vent protein adsorption. In other strategies, the internal In capillary gel electrophoresis, separation takes
wall of the capillary is coated with a polymer, cova- place inside a capillary filled with a gel that acts as a
lently bonded to the silica, that prevents interaction molecular sieve. Molecules with similar charge to mass
between the proteins and the negatively charged silica ratios are separated according to molecular size since
surface. For this purpose, ready-to-use capillaries with smaller molecules move more freely through the net-
coatings consisting of neutral-hydrophilic, cationic and work of the gel and therefore migrate faster than larger
anionic polymers are available. molecules. Different biological macromolecules (for
example, proteins and DNA fragments), which often
Electrolytic solution parameters have similar charge to mass ratios, can thus be separat-
(1) Buffer type and concentration Suitable buffers ed according to their molecular mass by capillary gel
for capillary electrophoresis have an appropriate buffer electrophoresis.
capacity in the pH range of choice and low mobility to
minimize current generation. Matching buffer-ion mo- Characteristics of Gels
bility to solute mobility, whenever possible, is im- Two types of gels are used in capillary electropho-
portant for minimizing band distortion. The type of resis: permanently coated gels and dynamically coated
sample solvent used is also important to achieve on- gels. Permanently coated gels, such as cross-linked
column sample focusing, which increases separation polyacrylamide, are prepared inside the capillary by
efficiency and improves detection. An increase in buff- polymerization of the monomers. They are usually
er concentration (for a given pH) decreases electro- bonded to the fused silica wall and cannot be removed
osmotic flow and solute velocity. without destroying the capillary. If the gels are used for
(2) Buffer pH The pH of the buffer can affect sepa- protein analysis under reducing conditions, the separa-
ration by modifying the charge of the analyte or addi- tion buffer usually contains sodium dodecyl sulfate and
tives, and by changing the electro-osmotic flow. In the samples are denatured by heating a mixture of so-
protein and peptide separation, changing the pH of the dium dodecyl sulfate and 2-mercaptoethanol or
buffer from above to below the isoelectric point (pI) dithiothreitol before injection. When non-reducing
changes the net charge of the solute from negative to conditions are used (for example, analysis of an intact
positive. An increase in the buffer pH generally in- antibody), 2-mercaptoethanol and dithiothreitol are not
creases the electro-osmotic flow. used. Separation in cross-linked gels can be optimized
(3) Organic solvents Organic modifiers (methanol, by modifying the separation buffer (as indicated in the
acetonitrile, etc.) may be added to the aqueous buffer capillary zone electrophoresis section) and controlling
to increase the solubility of the solute or other additives the gel porosity during the gel preparation. For cross-
and/or to affect the degree of ionization of the sample linked polyacrylamide gels, the porosity can be modi-
components. The addition of these organic modifiers to fied by changing the concentration of acrylamide
the buffer generally causes a decrease in the electro- and/or the proportion of cross -linker. As a rule, a de-
osmotic flow. crease in the porosity of the gel leads to a decrease in
(4) Additives for chiral separations For the separa- the mobility of the solutes. Due to the rigidity of these
tion of optical isomers, a chiral selector is added to the gels, only electrokinetic injection can be used.
separation buffer. The most commonly used chiral se- Dynamically coated gels are hydrophilic polymers,
lectors are cyclodextrins, but crown ethers, polysaccha- such as linear polyacrylamide, cellulose derivatives,
rides and proteins may also be used. Since chiral dextran, etc., which can be dissolved in aqueous sepa-
recognition is governed by the different interactions ration buffers giving rise to a separation medium that
between the chiral selector and each of the enantiomers, also acts as a molecular sieve. These separation media
the resolution achieved for the chiral compounds de- are easier to prepare than cross-linked polymers. They
pends largely on the type of chiral selector used. In this can be prepared in a vial and filled by pressure in a
regard, for the development of a given separation, it wall-coated capillary with no electroosmotic flow. Re-
may be useful to test cyclodextrins having a different placing the gel before every injection generally im-
cavity size (-, -, or -cyclodextrin) or modified proves the separation reproducibility. The porosity of
cyclodextrins with neutral (methyl, ethyl, hydroxyalkyl, the gels can be increased by using polymers of higher
etc.) or ionizable (aminomethyl, carboxymethyl, molecular mass (at a given polymer concentration) or
sulfobutyl ether, etc.) groups. When using modified by decreasing the polymer concentration (for a given
cyclodextrins, batch-to-batch variations in the degree polymer molecular mass). A reduction in the gel poros-
of substitution of the cyclodextrins must be taken into ity leads to a decrease in the mobility of the solute for
account since it will influence the selectivity. Other the same buffer. Since the dissolution of these poly-
factors controlling the resolution in chiral separations mers in the buffer gives low viscosity solutions, both
are concentration of chiral selector, composition and hydrodynamic and electrokinetic injection techniques
pH of the buffer and temperature. The use of organic can be used.
additives, such as methanol or urea can also modify the
resolution achieved. 3. Capillary Isoelectric Focusing
KP X 1821

In isoelectric focusing, the molecules migrate un- (1) Voltage Capillary isoelectric focusing utilizes
der the influence of the electric field, so long as they very high electric fields, 300 V/cm to 1000 V/cm in the
are charged, in a pH gradient generated by ampholytes focusing step.
having pI values in a wide range (poly- (2) Capillary The electro-osmotic flow must be re-
aminocarboxylic acids), dissolved in the separation duced or suppressed depending on the mobilization
buffer. strategy (see above). Coated capillaries tend to reduce
The three basic steps of isoelectric focusing are the electro-osmotic flow.
loading, focusing and mobilization. (3) Solutions The anode buffer reservoir is filled
(1) Loading step: Two methods may be employed: with a solution with a pH lower than the pI of the most
(i) loading in one step: the sample is mixed with acidic ampholyte and the cathode reservoir is filled
ampholytes and introduced into the capillary either with a solution with a pH higher than the pI of the most
by pressure or vacuum; basic ampholyte. Phosphoric acid for the anode and
(ii) sequential loading: a leading buffer, then the sodium hydroxide for the cathode are frequently used.
ampholytes, then the sample mixed with ampholytes, Addition of a polymer, such as methylcellulose, in
again ampholytes alone and finally the terminating the ampholyte solution tends to suppress convective
buffer are introduced into the capillary. The volume forces (if any) and electro-osmotic flow by increasing
of the sample must be small enough not to modify the viscosity. Commercial ampholytes are available
the pH gradient. covering many pH ranges and may be mixed if neces-
(2) Focusing step: When the voltage is applied, sary to obtain an expanded pH range. Broad pH ranges
ampholytes migrate toward the cathode or the anode, are used to estimate the isoelectric point whereas nar-
according to their net charge, thus creating a pH gradi- rower ranges are employed to improve accuracy. Cali-
ent from anode (lower pH) to cathode (higher pH). bration can be done by correlating migration time with
During this step the components to be separated mi- isoelectric point for a series of protein markers.
grate until they reach a pH corresponding to their isoe- During the focusing step precipitation of proteins
lectric point (pI) and the current drops to very low val- at their isoelectric point can be prevented, if necessary,
ues. using buffer additives such as glycerol, surfactants,
(3) Mobilization step: If mobilization is required for urea or zwitterionic buffers. However, depending on
detection, use one of the following methods. Three the concentration, urea denatures proteins.
methods are available:
(i) in the first method, mobilization is accomplished 4. Micellar Electrokinetic Chromatography (MEKC)
during the focusing step under the effect of the elec- In micellar electrokinetic chromatography, separa-
tro-osmotic flow; the electro-osmotic flow must be tion takes place in an electrolyte solution which con-
small enough to allow the focusing of the compo- tains a surfactant at a concentration above the critical
nents; micellar concentration (CMC). The solute molecules
(ii) in the second method, mobilization is accom- are distributed between the aqueous buffer and the
plished by applying positive pressure after the focus- pseudo-stationary phase composed of micelles, accord-
ing step; ing to the partition coefficient of the solute. The tech-
(iii) in the third method, mobilization is achieved after nique can therefore be considered as a hybrid of elec-
the focusing step by adding salts to the cathode res- trophoresis and chromatography. MECK is a technique
ervoir or the anode reservoir (depending on the di- that can be used for the separation of both neutral and
rection chosen for mobilization) in order to alter the charged solutes, maintaining the efficiency, speed and
pH in the capillary when the voltage is applied. As instrumental suitability of capillary electrophoresis.
the pH is changed, the proteins and ampholytes are One of the most widely used surfactants in MEKC is
mobilized in the direction of the reservoir which the anionic surfactant sodium dodecyl sulfate, although
contains the added salts and pass the detector. The other surfactants, for example cationic surfactants such
separation achieved, expressed as ΔpI, depends on as cetyltrimethylammonium salts, are also used.
the pH gradient (dpH/dx), the number of ampholytes The separation mechanism in MECK is as follows.
having different pI values, the molecular diffusion At neutral and alkaline pH, a strong electro-osmotic
coefficient (D), the intensity of the electric field (E) flow is generated and moves the separation buffer ions
and the variation of the electrophoretic mobility of in the direction of the cathode. If sodium dodecyl sul-
the analyte with the pH (-dµ/dpH): fate is employed as the surfactant, the electrophoretic
migration of the anionic micelle is in the opposite di-
D  (dpH / dx) rection, towards the anode. As a result, the overall mi-
pI  3  celle migration velocity is slowed down compared to
E  (d / dpH ) the bulk flow of the electrolytic solution.
In the case of neutral solutes, since the analyte can
Optimization partition between the micelle and the aqueous buffer,
The main parameters to be considered in the de- and has no electrophoretic mobility, the analyte migra-
velopment of separations are: tion velocity will depend only on the partition coeffi-
cient between the micelle and the aqueous buffer. In
1822 General Information

the electropherogram, the peaks corresponding to each cause excessive heat production that gives rise to tem-
uncharged solute are always between that of the perature gradients and viscosity gradients of the buffer
electroosmotic flow marker and that of the micelle (the in the cross-section of the capillary. This effect can be
time elapsed between these two peaks is called the sep- significant with high conductivity buffers such as those
aration window). For electrically charged solutes, the containing micelles. Poor heat dissipation causes band
migration velocity depends on both the partition coef- broadening and decreases resolution.
ficient of the solute between the micelle and the aque- (2) Temperature Variations in capillary temperature
ous buffer, and on the electrophoretic mobility of the affect the partition coefficient of the solute between the
solute in the absence of micelle. buffer and the micelles, the critical micellar concentra-
Since the mechanism in MEKC of neutral or tion and the viscosity of the buffer. These parameters
weakly ionized solutes is essentially chromatographic, contribute to the migration time of the solutes. The use
migration of the solute and resolution can be rational- of a good cooling system improves the reproducibility
ized in terms of the retention factor of the solute (k’), of the migration time for the solutes.
also referred to as mass distribution ratio (Dm), which (3) Capillary As in capillary zone electrophoresis,
is the ratio of the number of moles of solute in the mi- the dimensions of the capillary (length and internal
celle to those in the mobile phase. diameter) contribute to analysis time and efficiency of
separations. Increasing both effective length and total
t R  t0 V length can decrease the electric fields (working at con-
k' K S stant voltage), increase migration time and improve the
 tR  VM
t0  1   separation efficiency. The internal diameter controls
 tmc  heat dissipation (for a given buffer and electric field)
and consequently the sample zone diffusion.
tR: migration time of the solute ,
t0: analysis time of an unretained solute (determined Electrolytic solution parameters
by injecting an electro-osmotic flow marker (1) Surfactant type and concentration The type of
which does not enter the micelle, for instance surfactant, in the same way as the stationary phase in
methanol), chromatography, affects the resolution since it modi-
tmc: micelle migration time (measured by injecting a fies separation selectivity. Also, the log k’ of a neutral
micelle marker, such as Sudan III, which mi- compound increases linearly with the concentration of
grates while continuously associated in the mi- surfactant in the mobile phase.
celle), Since resolution in MEKC reaches a maximum when k’
K: partition coefficient of the solute, approaches the value of t m t0 , modifying the concen-
VS: volume of the micellar phase, tration of surfactant in the mobile phase changes the
VM: volume of the mobile phase. resolution obtained.
(2) Buffer pH Although pH does not modify the
Likewise, the resolution between two closely migrating partition coefficient of non-ionized solutes, it can mod-
solutes (RS) is given by the following equation: ify the electroosmotic flow in uncoated capillaries. A
decrease in the buffer pH decreases the electro-osmotic
t
1 ( 0 ) flow and therefore increases the resolution of the neu-
N   1 k 'b tmc tral solutes in MEKC, resulting in a longer analysis
RS    
4  k 'b 1 1  ( t0 )  k ' time.
a (3) Organic solvents To improve MEKC separation
tmc
of hydrophobic compounds, organic modifiers, such as
methanol, propanol, acetonitrile, etc., can be added to
N: number of theoretical plates for one of the so-
the electrolytic solution. The addition of these modifi-
lutes,
ers usually decreases migration time and the selectivity
: selectivity,
of the separation. Since the addition of organic modifi-
k’a, k’b: retention factors for both solutes, respec-
ers affects the critical micellar concentration, a given
tively (k’b > k’a)
surfactant concentration can be used only within a cer-
tain percentage of organic modifier before the
Similar, but not identical, equations give k’ and RS
micellization is inhibited or adversely affected, result-
values for electrically charged solutes.
ing in the absence of micelles and, therefore, in the
absence of partition. The dissociation of micelles in the
Optimization
presence of a high content of organic solvent does not
The main parameters to be considered in the de-
always mean that the separation will no longer be pos-
velopment of separations by MEKC are instrumental
sible; in some cases the hydrophobic interaction be-
and electrolytic solution parameters.
tween the ionic surfactant monomer and the neutral
solutes forms solvophilic complexes that can be sepa-
Instrumental parameters
rated electrophoretically.
(1) Voltage Separation time is inversely proportional
to applied voltage. However, an increase in voltage can
KP X 1823

(4) Additives for chiral separations For the separa- Apparent Number of Theoretical Plates
tion of enantiomers using MEKC, a chiral selector is
included in the micellar system, either covalently 2
t 
bound to the surfactant or added to the micellar separa- N  5.54   R 
tion electrolyte. Micelles that have a moiety with chiral  wh 
discrimination properties include salts of N- tR: migration time or distance along the baseline
dodecanoyl-L -amino acids, bile salts, etc. Chiral reso- from the point of injection to the perpendicular
lution can also be achieved using chiral discriminators, dropped from the maximum of the peak corre-
such as cyclodextrins, added to the electrolytic solu- sponding to the component,
tions which contain micellized achiral surfactants. wh: width of the peak at half -height.
(5) Other additives Several strategies can be carried
out to modify selectivity, by adding chemicals to the Resolution
buffer. The addition of several types of cyclodextrins The resolution (RS) between peaks of similar
to the buffer can also be used to reduce the interaction height of two components may be calculated using the
of hydrophobic solutes with the micelle, thus increas- following expression:
ing the selectivity for this type of compound.
The addition of substances, capable of modifying
 1.18  (tR 2  t R1 ) 
solute-micelle interactions by adsorption on the latter, RS    t R 2  t R1

is used to improve the selectivity of the separations in  wh1  wh 2 
MEKC. These additives may be a second surfactant tR1, tR2: migration times or distances along the base-
(ionic or non-ionic) which gives rise to mixed micelles line from the point of injection to the perpendicu-
or metallic cations which dissolve in the micelle and lars dropped from the maxima of two adjacent
form co-ordination complexes with the solutes. peaks,
wh1, wh2: peak widths at half -height.
Quantification When appropriate, the resolution may be calculat-
Peak areas must be divided by the corresponding ed by measuring the height of the valley (Hv) between
migration time to give the corrected area in order to: two partly resolved peaks in a standard preparation and
(1) compensate for the shift in migration time from run the height of the smaller peak (Hp) and calculating the
to run, thus reducing the variation of the response, peak-to-valley ratio:
(2) compensate for the different responses of sample
constituents with different migration times. Hp
Where an internal standard is used, verify that no p/v 
Hv
peak of the substance to be examined is masked by that
of the internal standard.
Symmetry Factor
Calculations The symmetry factor (AS) of a peak may be calcu-
From the values obtained, calculate the content of lated using the following expression:
the component or components being examined. When
prescribed, the percentage content of one or more w0.05
AS 
components of the sample to be examined is calculated 2d
by determining the corrected area(s) of the peak(s) as a w0.05: width of the peak at 1/20 of the peak height,
percentage of the total of the corrected areas of all d: distance between the perpendicular dropped from
peaks, excluding those due to solvents or any added the peak maximum and the leading edge of the
reagents. The use of an automatic integration system peak at 1/20 of the peak height .
(integrator or data acquisition and processing system)
is recommended. Tests for area repeatability (standard deviation of
areas or of the area/migration-time ratio) and for mi-
System Suitability gration time repeatability (standard deviation of migra-
In order to check the behavior of the capillary tion time) are introduced as suitability parameters. Mi-
electrophoresis system, system suitability parameters gration time repeatability provides a test for the suita-
are used. The choice of these parameters depends on bility of the capillary washing procedures. An alterna-
the mode of capillary electrophoresis used. They are: tive practice to avoid the lack of repeatability of the
retention factor (k’, only for micellar electrokinetic migration time is to use migration time relative to an
chromatography), apparent number of theoretical plates internal standard.
(N), symmetry factor (AS) and resolution (RS). In previ- A test for the verification of the signal-to-noise ra-
ous sections, the theoretical expressions for N and RS tio for a standard preparation (or the determination of
have been described, but more practical equations that the limit of quantification) may also be useful for the
allow these parameters to be calculated from the determination of related substances.
electropherograms are given below.
Signal-to-noise Ratio
1824 General Information

The detection limit and quantification limit corre- read the unsettled apparent volume to the nearest grad-
spond to signal-to-noise ratios of 3 and 10 respectively. uated unit and calculate the bulk density ρB by the fol-
The signal-to-noise ratio (S/N) is calculated using the lowing formula:
following expression:
M
B 
2 H V0
S/N 
h
H: height of the peak corresponding to the compo-  B : Bulk density by constant mass method
nent concerned, in the electropherogram obtained
with the prescribed reference solution, measured (g/mL)
from the maximum of the peak to the extrapolat- M : Mass of powder sample (g)
ed baseline of the signal observed over a distance V0 : Apparent volume of powder sample (mL)
equal to twenty times the width at half height,
h: range of the background in an electropherogram Record the average of 3 determinations and regard
obtained after injection of a blank, observed over the average value as the bulk density according to the
a distance equal to twenty times the width at the constant mass method. If a 30 g sample is too large to
half height of the peak in the electropherogram determine, adjust the mass of sample so as to provide
obtained with the prescribed reference solution an apparent volume of 60-100 mL.
and, if possible, situated equally around the place
where this peak would be found . Method 2 Constant volume method
Unless otherwise specified, pass a quantity of
sample sufficient to complete the test through a sieve
2. Determination of Bulk and No. 16 (1000 µm) to break up agglomerates that may
have formed during storage. Allow an excess of sample
Tapped Densities powder to pour into the measuring vessel having the
volume of V and mass of M0. Then, carefully scrape
Determination of Bulk and Tapped Densities is a excess powder from the top of the vessel using the
method to determine the bulk densities of powdered edge of a slide glass or other tool by smoothly moving
drugs under loose and tapped packing conditions, re- across it. Remove any material from the sides of the
spectively. Loose packing is defined as the state ob- vessel, and determine the total mass Mt and calculate
tained by pouring a powder sample into a vessel with- the bulk density ρB by the following formula:
out any consolidation, and tapped packing is defined as
the state obtained when the vessel containing the pow- (M t  M 0 )
der sample is to be repeatedly dropped a specified dis- B 
V
tance at a constant drop rate until the apparent volume
of sample in the vessel becomes almost constant. The
ρB: Bulk density by constant volume method (g/mL)
bulk density is expressed in mass per unit apparent
MT: Total mass of powder and measuring vessel (g)
volume of powder (g/mL). Because the bulk density is
M0: Mass of measuring vessel (g)
one of the measures of packing properties, compressi-
V: Volume of measuring vessel (mL)
bility and flow properties, and is dependent on the ‘his-
tory’ of the powder, it is essential to document the bulk
Record the average of 3 determinations and regard
density to specify how the determination was made.
the average value as the bulk density according to the
constant volume method.
Bulk density
The bulk density is an apparent density obtained
by pouring a powder sample into a vessel without any
consolidation. The determination of bulk density is
achieved by measuring the apparent volume of a pow-
der sample having a known mass in a graduated cylin-
der (Method 1) or by measuring the mass of powder in
a vessel having a known volume (Method 2).

Method 1 Constant mass method


Unless otherwise specified, pass a quantity of
sample sufficient to complete the test through a sieve
No. 16 (1000 m) to break up agglomerates that may
have formed during storage. Weigh accurately about 30
g of test sample, and pour it into a dry 100 mL gradu- Measuring vessel
ated glass cylinder (readable to 1 mL). Carefully level
the powder without consolidation, if necessary, and
KP X 1825

ρT: Tapped density by constant mass method (g/mL)


M: Mass of powder sample (g)
Vf: Final apparent volume of sample after tapping
(mL)

Record the average of 3 determinations and regard the


average value as the tap density according to the con-
stant mass method.

Method 2 Constant volume method


Unless otherwise specified, pass a quantity of
sample sufficient to complete the test through a sieve
No. 16 (1000 µm) to break up agglomerates that may
Supplementary cylinder have formed during storage. Attach a supplementary
cylinder to the stainless steel measuring vessel having a
The figures are in mm. known mass of M0 and a volume of V (Fig. 1), and then
This figure is one of the representations of 100 mL pour an excess of the sample into the vessel. After set-
vessel and supplementary cylinder. ting up the vessel in an adequate tapping apparatus
with a fixed drop height, carry out tapping at the rate
Figure 1. Vessels for the determination of bulk density and cumulative tap number specified for each appa-
by constant volume method and for the determination ratus. Then remove the supplementary cylinder from
tapped density the vessel and carefully scrape excess powder from the
top of the vessel by smoothly moving across it the edge
Tapped density of a slide glass or other tool. Remove any material
Tapped density is an apparent density obtained by from the sides of the vessel, and determine the total
mechanically tapping a measuring vessel containing a mass Mt. Calculate the tapped density ρT by the formu-
powder sample. The determination of tapped density is la:
achieved by measuring the apparent volume of a pow-
der sample having a known mass in a vessel after tap- (M t  M 0 )
ping (Method 1) or by measuring the mass of powder T 
V
in a vessel having a known volume after tapping
(Method 2).
ρT: Tapped density by constant volume method
(g/mL)
Method 1 Constant mass method Mt: Total mass of powder and measuring vessel (g)
Unless otherwise specified, pass a quantity of
M0: Mass of measuring vessel (g)
sample sufficient to complete the test through a sieve
V: Volume of measuring vessel (mL)
No. 16 (1000 µm) or a sieve No. 22 (710 µm) to break
up agglomerates that may have formed during storage.
Determine 3 measurements and calculate the av-
Weigh accurately about 100 g of test sample, and pour
erage and the relative standard deviation using 3 differ-
it into a 250 mL graduated glass cylinder (readable to 2
ent powder samples. Regard the average value as the
mL) without consolidation. If the sample is not suffi-
tap density according to the constant volume method.
cient, proceed according to the same procedure as that
If the relative standard deviation is not less than 2%,
described above by using a 100 mL graduated glass
repeat the test with further tapping.
cylinder (readable to 1 mL). It is essential to select
appropriate masses of the cylinder support, holder and
Note: Use balances readable to the nearest 0.1 g.
cylinder so as to ensure the dynamic stability of the
apparatus during tapping.
After attaching the glass cylinder containing the
powder sample to the tapping apparatus, carry out tap- 3. Disinfection and Sterilization
ping under the measuring conditions (tapping rate and
drop height) specified for each apparatus.
Methods
Unless otherwise specified, repeat increments of
50 taps or 1 minute until the difference between suc- Disinfection and Sterilization Methods are applied
ceeding measurements is less than 2%, and determine to kill microorganisms in processing equip-
the final apparent volume, Vf and calculate the tapped ment/utensils and areas used for drug manufacturing,
density ρT by the following formula: as well as to perform microbiological tests specified in
the monographs, and so differ from “Terminal Sterili-
M zation” and “Filtration Method” described in “Terminal
T  Sterilization and Sterilization Indicators”. The killing
Vf effect on microorganisms or the estimated level of ste-
rility assurance is greatly variable, so the conditions for
1826 General Information

disinfection and sterilization treatment must be chosen Microorganisms are killed by putting the object in boil-
appropriately for each application. Generally, the fol- ing water. This method is used for a product which
lowing methods are to be used singly or in combination may be denatured by the moist heat method. As a rule,
after appropriate optimization of operation procedures the product is put in boiling water for 15 minutes or
and conditions, in accordance with the kind and the more.
degree of the contaminating microorganisms and the (iii) Intermittent method
nature of the items to which the methods are applied. Microorganisms are killed by heating for 30 to 60
The validation of sterilization in accordance with minutes repeatedly, three to five times, once a day in
Terminal Sterilization and Sterilization Indicators is water at 80 to 100 C or in steam. This method is used
required when the methods are applied to the manufac- for a product which may be denatured by the moist
turing process of drug products. heat method. There is another method called the low
temperature intermittent method with repeated heating
1. Disinfection methods at 60 to 80 C. During the intermission periods be-
These methods are used to reduce the number of living tween heating or warming, a suitable temperature for
microorganisms, but do not always remove or kill all the growth of microorganisms of 20 C or higher, must
microorganisms present. Generally, disinfection is be maintained.
classified into chemical disinfection with the use of (iv) Ultraviolet method
chemical drugs (disinfectants) and physical disinfec- As a rule, microorganisms are killed by irradiation with
tion with the use of moist heat, ultraviolet light, and ultraviolet rays as a wavelength of around 254 nm.
other agents. This method is used for products which are resistant to
ultraviolet rays, such as smooth-surfaced articles, facil-
1.1. Chemical disinfection ities, and equipment, or water and air. This method
Microorganisms are killed with chemical drugs. The does not suffer from the occurrence of resistance,
killing effect and mechanisms of a chemical drug differ which is observed in chemical disinfection, and shows
depending on the type, applied concentration, action a killing effect on bacteria, fungi, and viruses. It must
temperature, and action time of the chemical drug used, be taken into consideration that direct ultraviolet irra-
the degree of contamination on the object to be disin- diation of the human body can injure the eyes and skin.
fected, and the series and state (e.g., vegetative bacteria
or spore bacteria) of microorganisms. 2. Sterilization methods
Therefore, in applying the method, full consideration is 2.1. Heating methods
required of the sterility and permissible storage period In these methods, the heating time before the tempera-
of the prepared chemical drug, the possibility of re- ture or pressure reaches the prescribed value differs
sistance of microorganisms at the site of application, according to the properties of the product, the size of
and the effect of residual chemical drug on the product. the container, and the conditions. The duration of heat-
In selecting a suitable chemical drug, the following ing in conducting these methods is counted from the
items should be considered in relation to the intended time when all the parts of the product have reached the
use. prescribed temperature.
1) The antimicrobial spectrum (i) Moist heat method
2) Action time for killing microorganisms Microorganisms are killed in saturated steam at a suit-
3) Action durability able temperature and pressure. This method is general-
4) Effect of the presence of proteins ly used for heat-stable substances, such as glass, porce-
5) Influence on the human body lain, metal, rubber, plastics, paper, and fiber, as well as
6) Solubility in water heat-stable liquids, such as water, culture media, rea-
7) Influence on the object to be disinfected gents, test solutions, liquid samples, etc. As a rule, one
8) Odor of the following conditions is used.
9) Convenience of use
10) Easy disposability 115 ~ 118 C for 30 minutes
11) Influence on the environment at disposal 121 ~ 124 C for 15 minutes
12) Frequency of occurrence of resistance 126 ~ 129 C for 10 minutes
1.2. Physical disinfection (ii) Dry-heat method
Microorganisms are killed without a chemical drug. Microorganisms are killed in dry-heated air. This
(i) Steam flow method method is generally used for heat-stable substances,
Microorganisms are killed by direct application of such as glass, porcelain, and metal, as well as heat-
steam. This method is used for a product which may be stable products, such as mineral oils, fats and oils,
denatured by the moist heat method. As a rule, the powder samples, etc. This method is generally con-
product is kept in flowing steam at 100 C for 30 to ducted in the way of direct heating by gas or electricity
60 minutes. or circulating heated air. As a rule, one of the following
(ii) Boiling method conditions is used.
KP X 1827

160 ~ 170 C for 120 minutes ucts.


170 ~ 180 C for 60 minutes
180 ~ 190 C for 30 minutes 2. Significance of Dissolution Testing
Drug absorption from a solid dosage form, such as
2.2. Irradiation methods the tablet and the capsule, after oral administration is
(i) Radiation method governed by a number of factors, including the release
Microorganisms are killed by gamma-rays emitted properties of the drug substance from the drug product,
from a radioisotope or electron beam and bremsstrah- the dissolution or solubilization properties of the drug
lung (X-ray) generated from an electron accelerator. under a given physiological condition and the permea-
This method is generally used for radiation-resistant bility across the intestinal epithelial barrier. A dissolu-
substances such as glass, porcelain, metal, rubber, plas- tion testing is a test that determines not only the release
tics, fiber, etc. The dose is decided according to the of the drug substance but also the dissolution or
material properties, and the degree of contamination of solubilization of the drug. Ultimately, a dissolution
the product to be sterilized. Special consideration is testing can lead to the estimation of the performance of
necessary of the possibility of qualitative change of the the drug product in the biological system. Therefore,
product after the application of the method. when a dissolution testing is undertaken, it is advisable
(ii) Microwave method to consider the solubility of the drug, the dissolution,
Microorganisms are killed by the heat generated by the intestinal permeability, the characteristics of the
direct microwave irradiation. This method is generally dosage form, the pharmacokinetic properties as well as
used for microwave-resistant products such as water, physiological factors. In order to mimic physiological
culture media, test solutions, etc. As a rule, microwave conditions after the oral administration, the conditions
radiation with a wavelength of around 2450±50 MHz is for dissolution testing vary, e.g., a various dissolution
used. media with varying pHs to represent the environments
in the gastrointestinal tract, a number of settings in the
2.3. Gas methods rotational speed to simulate the peristaltic movement of
Microorganisms are killed by a sterilizing gas. Suitable the gastrointestinal tract and addition of lipids, en-
gases for killing microorganisms include ethylene ox- zymes and/or surfactants to mimic the physiological
ide gas, formaldehyde gas, hydrogen peroxide gas, conditions in the tract. Sometimes the results from a
chlorine dioxide gas, etc. Temperature, humidity, the dissolution test may be used to predict the kinetics of
concentration of gas, and the exposure time differ in the absorption of the drug (viz, in vitro-in vivo correla-
accordance with the species of gas used. As sterilizing tion). However, it is noted that the condition of dissolu-
gases are generally toxic to humans, full consideration tion testing does not always attempt to represent com-
is required of the environmental control for the use of pletely physiological conditions in the gastrointestinal
gases and the concentration of residual gas. In some of tract. That is, useful information on the quality control
the gas methods, it may be difficult to measure or esti- and the quality assurance of a given drug product can
mate quantitatively the killing of microorganisms. also be obtained from the dissolution test that does not
completely represent physiological conditions in hu-
2.4. Filtration method man gastrointestinal tract.
Microorganisms are removed by filtration with a suita- A. Lot to lot uniformity in the quality
ble filtering device. This method is generally used for B. Development of composition of drug product and
gas, water, or culture media and test solutions contain- of formulation
ing a substance that is water-soluble and unstable to C. Understanding release mechanism of drug from
heat. As a rule a filter having a pore size of 0.22 m or product
smaller is used for the sterilization. However, in this D. Determination of pharmaceutical equivalence dur-
method, a filter with a pore size of 0.45 m of smaller ing storage
is permitted to be used. E. Determination of pharmaceutical equivalence of
post approval changes in drug substance, in com-
position, and in method, site and scale of manufac-
ture
4. Guideline for Setting
Dissolution Specification of Oral 3. Glossary
a. Conventional release dosage forms (also known as
Dosage Forms immediate release dosage forms)
A conventional release dosage form or immediate
1. Objective release dosage form is a dosage form in which the
This guideline is intended to provide specified ap- dissolution profile is primarily governed by the
proaches for setting dissolution specification of oral physicochemical property of the drug substance
solid dosage forms such as tablets and capsules. By and cannot be intentionally altered simply by
securing quality uniformity of drug product, this guide- changing the amount of the drug substance in the
line contributes to production of qualified drug prod- product or the method of manufacture.
1828 General Information

b. Delayed release dosage forms (also known as en- rotary basket methods is recommended for cap-
teric coated dosage forms) sules, a dosage form that is not ease to sink, or
A delayed release dosage form or enteric coated for slowly dissolving dosage forms. Although
dosage form is a dosage form in which the dissolu- dissolutions from tablets are typically tested
tion of the drug is delayed for a certain period after with the paddle method, the rotary basket meth-
the administration so that the drug is not released od, rather than the paddle method, may be more
in the acidic condition of the stomach by means of suitable in case where the disintegrated debris
composing the product with specialized composi- are settled down at the bottom of the flask there-
tions or specialized methods. Pharmacokinetic cri- by causing a delay in the dissolution. Also disso-
teria of the dosage form are such that the dissolu- lution apparatuses for dissolution testing is rec-
tion profile of the dosage form is comparable to its ommended to accomplish the testing of Suitabil-
conventional release dosage form counterpart ex- ity test for dissolution apparatus (see Appendix 1)
cept that the release occurs after a certain time. for an every certain period of time.
c. Prolonged release dosage forms (also known as
extended release dosage forms)
A prolonged release dosage form or extended re-
lease dosage form is a dosage form in which the
release of the drug substance is prolonged, com-
pared with that from the conventional release dos-
age form of the same route of administration, and
the frequency of administration is reduced com-
pared with that in the conventional forms. These Apparatus 1 Apparatus 2
dosage forms are typically manufactured by means (basket) (paddle)
of specialized compositions and/or specialized
methods.

4. General consideration in setting the dissolution


specifications
a. Classification of dosage forms
In this guideline, dosage forms are classified into
conventional release dosage forms, delayed release
dosage forms and prolonged release dosage forms
as specified in section 3. Glossary. Apparatus 3 Apparatus 4
b. Apparatus for dissolution testing (flow-through cell) (reciprocating cylinder)
i. Apparatus
Currently, seven types of dissolution apparatuses
are specified in the official compendium and the
types are illustrated in Fig. 1. Depending on the
properties of the dosage form, a suitable appa-
ratus may be selected for the dissolution testing.
Two of the most frequently used methods are the
Method 1, the rotary basket method, and the
Method 2, the paddle method, both specified in
the Dissolution Test. The rotary basket method Apparatus 5 Apparatus 6
and the paddle method are both simple and ro- (paddle over disk) (cylinder)
bust testing procedure and are highly standard-
ized globally. Therefore, unless a special disso-
lution testing is required, the rotary basket
method and the paddle method are recommend-
ed. When a dissolution test is to be conducted by
the rotary basket method or the paddle method,
the apparatus should conform to the specifica-
tion of the Dissolution Test as defined in the
General Tests of the Korean Pharmacopoeia.
When a dissolution test is to be conducted for
the development of a new drug product, the se-
lection between the rotary basket method and
the paddle method depends on the consideration
of the in vitro dissolution pattern and in vivo
pharmacokinetic characteristics. In general, the
KP X 1829

tion of drug product, manufacturing process,


equipment, drug substance, etc. In general, a
dissolution medium that results in a very rapid
dissolution is not suitable for the identification
and, thus, it is advisable to select a dissolution
medium that shows a relatively slower rate of
dissolution.
Acryl rod Angled disk - In case where a given drug substance adsorbs
to the surface of dissolution apparatus, there-
by creating a problem in analyzing the results,
use a dissolution medium with a suitable
Teflon cylinder agent that prevents the adsorption.
- Suitable surfactants may be added to dissolu-
tion medium to enhance solubility of a very
poorly soluble drug. However, addition of or-
ganic solvent to medium is not recommended
for dissolution test.
- In case where the identical result cannot be
obtained for the test with the deaerated disso-
Spring holder Reciprocating disk lution medium and for that with the non-
deaerated dissolution medium, then the
Apparatus 7 (reciprocating holder) deaeration procedure is necessary (see Ap-
pendix II).
Figure 1. Types of Dissolution Apparatuses 2) Characteristics of dissolution medium
a) Water
Since water does not have any buffer capac-
ii. Rotational speed ity, the disadvantages of water as a dissolu-
In general, the rotational speed is set at 50 rpm tion medium include the potential changes
for the paddle method and at 100 rpm for the ro- in the pH and in the surface tension of the
tary basket method. A rotational speed faster medium by the dissolution of drug and/or
than 100 rpm is necessary for prolonged dosage the additives. Despite the disadvantages,
forms tested by the paddle method. However, it however, water is the most convenient me-
is not always necessary to set the above men- dium and ecologically-acceptable. There-
tioned speeds in a test. For example, in case fore, water is the most widely used dissolu-
where 100% of the drug is dissolved within 10 tion medium, in case where the dissolution
minutes using the rotary basket method at the pattern from the formulation is not affected
rotational speed of 100 rpm for a dissolution test, by using water as the medium.
it is advisable to set a lower rotation speed b) pH 1.2 Dissolution medium
and/or to change the composition of the dissolu- Use the disintegration 1st fluid. This medi-
tion medium so that the dissolution pattern is um is prepared using hydrochloric acid and
discernable. Therefore, the rotational speed of a sodium chloride, and the concentration of
test should be set at a speed that is able to identi- hydrochloric acid in the medium is set at
fy potential lack of pharmaceutical bioequiva- about 0.1 mol/L. In case where a particular
lence of a drug product. drug becomes unstable in the medium, it is
iii. Dissolution medium necessary that the testing condition is modi-
1) General considerations fied to maintain the stability. In fact, the
- Although the dissolution medium, having the dissolutions from a number of formulations
pH of 1.2, 4.0, 4.5, 6.8 or 7.4, is generally are tested with the media having the con-
used, the medium may be set at a different pH centration of hydrochloric acid ranging
and have a different composition, if necessary from 0.001 mol/L to 0.1 mol/L.
because of the property of the dosage form c) pH 4.0/4.5 Dissolution medium
(e.g., the stability and/or the solubility of the o pH 4.0 Dissolution medium: Use 0.05
drug) or of a specific experimental purpose. mol/L sodium acetate buffer solution [a
Under these circumstances, the properties, in- mixture of 0.05 mol/L acetic acid and
cluding the stability and solubility, of the drug 0.05 mol/L sodium acetate (82:18)].
substance must be considered. o pH 4.5 Dissolution medium: Use a buffer
- Select a dissolution medium that is able to solution prepared by dissolving 2.99 g of
properly identify the difference in the dissolu- sodium acetate trihydrate and 1.66 g of
tion rate between drug products or between glacial acetic acid in water to make 1000
lots according to the factors that are composi- mL.
1830 General Information

o In case where these buffer solutions in- medium is not the rate limiting step in the
teract with a drug to create undesirable dissolution rate by maintaining a sink con-
problem, a buffer solution prepared from dition in the test.
citrate or phosphate may be alternatively b) Use of surfactant
used. It is advisable to use a sufficient volume of
d) pH 6.0 Dissolution medium dissolution medium in the test when surfac-
Use disodium phosphate and citric acid tant is added to the medium to enhance the
buffer solution (pH 6.0) of the Korean solubility of a poorly water-soluble drug.
Pharmacopoeia. c) Ease of assay
e) pH 6.8 Dissolution medium For a dissolution test with a drug product
Use the disintegration 2nd fluid. having a trace amount of drug, it is recom-
f) Simulated gastric fluid mended to use the possible minimum in
Use simulated gastric fluid TS of USP. volume when the concentration of the dis-
g) Simulated intestinal fluid solved drug may be lower than the limit of
Use simulated intestinal fluid TS of USP. quantification thereby creating an assay
h) Neutral and basic dissolution medium problem.
Although the dissolution medium, having d) Temperature of test medium
the pH of 1.2, 4.0, 4.5, 6.8 or 7.4, is gener- In general, maintain the temperature of the
ally used, the medium may be set at a dif- medium at 37 ± 5 °C for the dissolution test
ferent pH and have a different composition, of oral solid dosage forms.
if necessary because of the property of the iv. Dissolution testing of drug product containing
dosage form (e.g., the stability and/or the poorly soluble drug
solubility of the drug) or of a specific exper- 1) Determination of impact of medium pH on
imental purpose, and the dissolution is test- dissolution rate
ed. Examples of the special conditions in- Conduct a series of dissolution tests with var-
clude the cases when a particular drug is the ious media such as water, pH 1.2 dissolution
most stable in basic dissolution media, medium, pH 4.0/4.5 dissolution medium and
when basic condition allows a discriminat- pH 6.8 dissolution medium. In these dissolu-
ing analysis or when a given drug has a suf- tion studies, use faster the rotational speed
ficient solubility in basic media. When a (for example, the rotational speed of 120 rpm
dissolution is tested for a product in basic for the rotary basket method and the rotational
condition, coupled with the HPLC assay us- speed of 75 rpm for the paddle method). Sam-
ing C18 column, it may be necessary to neu- ple a portion of the medium at suitable times
tralize or sufficiently dilute the test solution after the initiation of the test, calculate the rate
before the introduction to the column be- of dissolution for the drug and graphically
cause the packing material tends to be un- present the results. Based on these results, the
stable at high pHs. dissolution medium is selected for subsequent
3) Volume of dissolution medium tests. In case where the dissolution rate of the
Although the volume of dissolution medium is drug is below the range between 70 and 85%
typically 900 mL in the test, it may be adjust- of the drug, select a medium that shows the
ed to 500-1 L, depending on the testing condi- most rapid dissolution rate and consider addi-
tion. Dissolution test flask ranging from 150 tion of surfactants to the medium.
mL to over 1 L in size is currently available. 2) Selection of suitable surfactant
General considerations for setting the volume Interaction between drug and surfactant is
of dissolution medium are as follows; governed by the physicochemical properties
a) Solubility of drug of the drug and the surfactant. Therefore, for a
Volume of dissolution medium is sufficient given drug, a various surfactants, e.g., non-
enough to maintain a sink condition during ionic, anionic and cationic surfactants, have to
the testing. In general, a sink condition is a be screened for the selection. In this case, the
condition where at least three times (gener- concentration is recommended to set initially
ally 5-10 times) the volume for the com- at 2% and the suitability is tested for surfac-
plete dissolution of a given amount of drug tants such as;
is available. Therefore, under such condi- o Anionic surfactant: Sodium lauryl
tion, the concentration of the drug should sulfate (SLS)
always be less than 30% of the intrinsic sol- o Cationic surfactant: Cetyltrimethyl
ubility when the drug product is completely ammonium bromide (CTAB)
dissolved in the volume of medium used in o Non-ionic surfactant: Polysorbate 80,
the test. In the dissolution test, it is neces- 40 and 20, lauryl dimethylamine N-
sary to set the dissolution specification such oxide (LDAO)
that the process of the dissolution of drug to 3) Determination of surfactant concentration
KP X 1831

Use the lowest possible concentration of the vi. Two stage dissolution testing for gelatin cap-
surfactant that shows a sufficient dissolution sules
rate within the specified time of the comple- For the case of the dissolution test with hard- or
tion of the test (i.e., in 2 hours or 6 hours). To soft-gelatin capsules, a two-stage dissolution
accomplish this, study the dissolution with a testing is recommended. The first stage dissolu-
gradual decrease in the surfactant concentra- tion is tested using typical dissolution medium
tion such as from the initial 2%, then 1.5, 1.0, as described in the previous section. When the
0.75, 0.5 and finally to 0.25%. dissolution rate is too slow to be tested properly,
v. Dissolution testing for delayed release dosage proceed to the second stage dissolution test with
forms an addition of enzyme to the test medium. Pep-
To confirm the function of enteric coating or the sin or pancreatin is typically used as the diges-
diffusion control of the drug by coating, conduct tive enzyme for the test, and the suitable enzyme
a series of dissolution tests first in an acidic is selected depending on the pH of the medium.
condition (i.e., acid stage) and then in buffer (i.e., When water or a dissolution medium having the
buffer stage). pH of less than 6.8 is used in the test, pepsin is
As an example for a dissolution test for a de- added with the activity of pepsin being not more
layed release dosage form in the official com- than 750000 units per 1 L of the dissolution me-
pendium, dissolution may be tested as directed dium. In this case, the amount of pepsin addition
in the procedure for delayed release dosage is not more than 3.2 g per 1 L of the medium.
forms or first in 0.1 mol/L hydrochloric acid so- For a dissolution medium having the pH not less
lution, an acidic condition, and, then, in pH 6.8 than 6.8, pancreatin is used as the enzyme with
phosphate buffer in the buffer condition. For the the activity of pancreatin being not more than
dissolution testing via the two-stage approach, 1750 units per 1 L of the dissolution medium. In
there are next ways; this case, the amount of pancreatin addition is
1) Dissolution is first tested for 2 hours in 750 not more than 50 mg per 1 L of the medium. In
mL of 0.1 mol/L hydrochloric acid solution at case where water is used as the dissolution me-
37 ± 5 °C. After 2 hours, a portion of the dis- dium at the first stage, 0.1 mol/L hydrochloric
solution medium is sampled for the quantifi- acid solution containing pepsin or pH 6.8 phos-
cation of the drug in the medium. To the phate buffer containing pancreatin may be used
above mentioned 0.1 mol/L hydrochloric acid as the second stage medium. However, if a high
dissolution medium, add 250 mL of 0.2 mol/L concentration of surfactant, such as sodium
sodium phosphate solution, previously laury sulfate, is used in the first stage medium, a
warmed to 37 ± 5 °C. If necessary, the pH of care should be taken in the second stage test be-
the medium may be adjusted with 2 mol/L hy- cause the presence of surfactant may cause de-
drochloric acid or with 1 mol/L sodium hy- naturation of the enzymes in the medium.
droxide, and the dissolution further tested (the
adjustment of the pH should be carried out 5. Dissolution Specification
within not more than 5 minutes of the addition a. Conventional release dosage forms
of 0.2 mol/L sodium phosphate solution). There are two approaches in setting the dissolution
2) Dissolution is first tested for 2 hours in 1 L specification for conventional release dosage
of 0.1 mol/L hydrochloric acid solution at 37 forms.
± 5 °C. After 2 hours, a portion of the dissolu- i. Singe-point specifications: This approach is typ-
tion medium is sampled and the remaining ically applicable to drug products containing
medium is discarded. To the dissolution flask, highly soluble drug substances
transfer 1 L of pH 6.8 phosphate buffer and (Biopharmaceutics Classification System(BCS)
perform the Dissolution Test (prepared by Class I and III) in setting the dissolution specifi-
adding 750 mL of 0.1 mol/L hydrochloric acid cation for the purpose of routine quality control.
solution to 250 mL of 0.2 mol/L sodium phos- (e.g., ‘Perform the test according to the next, the
phate solution, and, if necessary, adjusting the dissolution rate of Acetaminophen in 30 minutes
pH to 6.8 by the addition of 2 mol/L hydro- should be not less than 80%.’) The single-point
chloric acid or of 1 mol/L sodium hydroxide), specification may be set for the dissolution test
previously warmed to 37 ± 5 °C. Another way of a drug product in which not less than 80% of
is to transfer the paddle or the rotary basket, the drug is dissolved within 20-30 minutes, even
containing the sample, from the apparatus of in the presence of the lag time (typically found
the first dissolution study with 0.1 mol/L hy- in film coated formulations and capsules), of the
drochloric acid solution to a new dissolution initiation of the test. It is not necessary to at-
flask containing pH 6.8 phosphate buffer and tempt to construct in vitro-in vivo correlation for
to continue to study the dissolution in the new such product in the development stage.
set up. ii. Two-point specifications: This approach is typi-
cally applicable to drug products containing
1832 General Information

poorly soluble drug substances (BCS Class II i. Number of test specimen


and IV) in setting the dissolution specification Proceed with 6 test specimens selected as di-
for the purpose of routine quality control. In rected in the section 6-a and perform the disso-
general, the two-point specification is required lution test.
for drug products having the dissolution time is ii. Interval of sampling of dissolution solution
not less than 45 minutes and is more useful than Sample the dissolution solution up to 2 hours of
the single-point specification. the test at appropriate intervals (e.g., typically at
b. Delayed release dosage forms 5, 10, 15, 30, 45, 60, 90 and 120 minutes) for
The dissolution specification is set at not more each dissolution medium. However, the test may
than 10% for the acid stage and at the level speci- be terminated at a point when the final rate of
fied in conventional release dosage forms for the dissolution reaches not less than 85%.
buffer stage. iii. Conditions for dissolution testing
c. Prolonged release dosage forms 1) Dissolution apparatus
Set the dissolution specification using at least three a) Use the paddle method in the Dissolution
time points. In this case, the sampling point should Test of the Korean Pharmacopoeia as a pre-
be such that the dissolution from the samples rep- ferred method.
resents the initial, middle and final stage of the b) Use other test method such as the rotary
dissolution. The average rate of dissolution at the basket method when, in the paddle method,
final stage should reach as close as possible to the disintegrated debris is settled down at
80%. the bottom to form aggregates thereby inter-
The initial stage time point (typically 1-2 hours of fering the test.
the initiation of the test) is selected so that the c) A sinker may be used when the test speci-
sample is able to identify the presence of initial men floats so that appropriately reproduci-
burst of dissolution from the drug product. In gen- ble results cannot be obtained.
eral, 20-30% of the drug should be dissolved in the 2) Dissolution medium
initial stage. The middle point is selected to identi- a) pH 1.2: Use the medium 1 of the dissolu-
fy the dissolution of 50% of the drug and the final tion test in the Korean Pharmacopoeia
point is selected to identify the dissolution of not b) pH 4.0/4.5: 0.05 mol/L sodium acetate
less than 80% of the drug thus confirming the dis- buffer solution*
solution of the majority of the drug by the final 0.05 mol/L sodium acetate buffer solution*:
sampling point. In case where the average rate of a mixture of 0.05 mol/L acetic acid and 0.05
dissolution does not reach 80% by the completion mol/L sodium acetate (82:28). But, the pH
of the test, however, the time of completion of the and the composition of the test medium
test is set at a point when the rate of dissolution may be modified depending on the pKa of a
does not change any more. given drug substance
For conventional release dosage forms, delayed re- c) pH 6.8: Use the medium 2 of the dissolu-
lease dosage forms and prolonged release dosage tion test in the Korean Pharmacopoeia
forms, the drug product should always conform to d) Water
the dissolution specification during the indicated e) Addition of surfactant to dissolution medi-
time of usage (i.e., within the expiration date). um: Use the medium in which the final rate
of dissolution is less than 85% in all medi-
6. Approaches in setting the dissolution specification um medium indicated in the section i)-iv) of
In general, the dissolution specification is set accord- 4-b.
ing to the following approach. However, in case 3) Rotational speed
where more appropriate method is available or other For the paddle method, initially use 50 rpm
suitable dissolution testing condition (e.g., the disso- and increase the speed to 75 rpm if the rate of
lution apparatus, the rotational speed and/or the dis- dissolution is slow. The rotational speed may
solution medium etc.) that better represents the char- be occasionally set at 100 rpm, but the rota-
acteristics of the dosage form, the other condition tional speed not less than 150 rpm is not used.
may be used in the test. iv. Selection of dissolution test conditions
a. Selection of test specimen 1) Conventional release dosage forms
i. For a drug, select the lot(or equivalent) that has Use the testing condition that results in 70-85%
been used for the clinical test or the bioavailabil- of the rate of the dissolution. Since the testing
ity test or the bioequivalence test. condition showing a rapid rate of dissolution
ii. Select the lot that the difference between the is not generally discriminating, select the dis-
content of test specimen and its labeled amount solution medium and the rotational speed that
is not more than 5% and difference between results in a relatively slow rate of dissolution
content of the test specimen and the content of (not more than 1 hour).
reference is not more than 5%. 2) Delayed release dosage forms
b. The first sage (a preliminary test) Proceed with the same testing condition in the
KP X 1833

section 6-b-iv)-1) and consult to the condi- of the drug is set as the initial point (1-2
tions in the section 5-b. In this case, use the hours), a point of about 50% dissolution of the
media of pH 1.2 and pH 6.8 as the test media. drug is set as the middle point, and a point of
3) Prolonged release dosage forms about 80% dissolution of the drug or a point
Select at least three time points for sampling when the rate of dissolution does not change
such that a point of about 20-30% dissolution any more is set as a the final point. The rates
of the drug is set as the initial point (1-2 of dissolution for the initial point and the
hours), a point of about 50% dissolution of the middle point are set to ‘the average rate of
drug is set as the middle point and a point of dissolution rate in ± 15%’ and the rate for the
about 80% dissolution of the drug is set as the final point being set to ‘the average rate of
final point. dissolution in – 10%’.
c. The second stage (the dissolution test) 3) Established test the dissolution according to
i. Proceed with the test conditions selected based the preliminary specification using the refer-
on the preliminary test and perform the test ence drug and verify the validity of the speci-
ii. Number of test specimen fication, including the analytical procedure.
Proceed with the test specimens from three lots
having 12 test specimens per each lot and per- 7. Preparation of validation data for dissolution test
form the test. a. Dissolution medium
iii. Dissolution specification Briefly summarize the reason for the selection of
1) Conventional release and delayed release the dissolution medium.
dosage forms b. Dissolution test procedure
a) Time span of test: For conventional release i. Describe the procedure for the dissolution test
dosage forms, use not more than 1 hour as and analysis according to the following items.
the time span of the test. For delayed re- o Dissolution apparatus
lease dosage forms, use not more than 2 o Preparation of the standard solution
hours in the acid stage and not more than 1 o Preparation of the test solution: Sampling
hour in the buffer stage. time of the dissolution solution, dilution, fil-
b) Setting specification of dissolution tration (if there is a filtration procedure in-
Set the specification of the dissolution for volved, present the data for the recovery
the test product at the value of about 10% evaluation)
less than the average rate of dissolution at o Analytical procedure for the quantification
the time point that reaches an almost plat- in the dissolution solution (e.g., HPLC etc.)
eau in the graphical representation of the re- o Formula for the calculation
sults for the dissolution test in the lot that o Dissolution specification
shows the medium rate of dissolution ii. Submit chromatograms or spectra of the blank
among the three lots. test solution, the standard solution and the test
 Single-point specification solution.
This specification is applicable to drug c. Validation
product containing a highly soluble drug Describe the validation data for the dissolution test
substance (e.g., BCS Class I or III). Set the and the analytical procedure.
specification as the lower boundary of the i. Data related to the stability of the test solution
dissolution rate at a time point that shows during the analysis
the rate in the range of 70-85%. ii. Analytical procedure
 Two-point specification o Perform the test according to the ‘Guideline
This specification is applicable to drug of Validation of Analytical Procedures for
product containing gently dissoluble drug Pharmaceuticals’.
substance (e.g., BCS Class II). 15 Minutes - Range
is generally used as the first point and 30, : Demonstrate linearity, accuracy and preci-
45 or 60 minutes as the second point for the sion within the specification of the dissolution ±
specification. For the drug product in which 20%.
a rapid dissolution may affect the efficacy (For example, for a delayed release dosage
or the adverse reaction, or which has a nar- form having the dissolution specification of
row therapeutic index (see the Attachment 2 not more than 20% at 1 hour, not less than
of the Minimum Requirements for Bioe- 90% at 9 hours, submit the data demonstrat-
quivalence Test), the specification is set us- ing the accuracy, precision and linearity in
ing the two-point testing (if necessary, set the range of 0-110% of the labeled amount).
both upper and lower boundary values).
2) Prolonged release dosage forms Appendix I
Select at least three time points for sampling
such that a point of about 20-30% dissolution Suitability test for dissolution apparatus
1834 General Information

Suitability test has to be conducted to determine concentration and use the standard solutions for
whether a frequently used dissolution apparatus can be the analysis. In case where the test solution is fil-
properly used and the test is carried out by an adequate tered, test for the potential adsorption of salicylic
test procedure. It is recommended that the suitability acid to the filter medium.
test be conducted typically twice a year. In addition,
the test has to be performed when the dissolution appa- Table 1. Dissolution specification of the salicylic acid
ratus is moved or a significant change has been made standard tablet (lot number O)
to the apparatus. Furthermore, the suitability test is Type of dissolution ap- Rate of dissolution at 30
required when the test method is modified from the paratus minutes of the dissolu-
rotary basket method to the paddle method and vice tion test (%)
versa. The suitability of a dissolution apparatus for the Apparatus 1 (the rotary 23-29
rotary basket method or for the paddle method is tested basket method)
using two standard calibrators (USP dissolution cali- Apparatus 2 (the paddle 17-26
brator: www.usp.org). The test is conducted using one method)
tablet of the non-dissolvable salicylic acid standard
tablet and one tablet of the dissolvable prednisone 2. Suitability test for dissolution apparatus using the
standard tablet. For the case of the salicylic acid tablet, 10 mg prednisone standard tablet (lot number N)
50 mmol/L phosphate buffer, pH 7.4 is used as the dis- a. Use 500 mL of water as the dissolution medi-
solution medium and, for the case of the prednisone um.
tablet, water is used as the medium. b. To deaerate the dissolution medium, heat the
1. Suitability test for dissolution apparatus using the solution to 41 °C while stirring, and filter the
300 mg salicylic acid standard tablet (lot number solution under vacuum using the filter paper
O) of the pore size of 0.45 µm. During the filtra-
a. Use 900 mL of 50 mmol/L phosphate buffer tion procedure, continue stirring the filtrate.
as the dissolution medium. In this case, the pH After the filtration procedure, cap the flask
of the buffer is adjusted to 7.4 ± 0.05 in the and continue the stirring for 5 additional
room temperature. minutes under vacuum.
b. To deaerate the dissolution medium, heat the c. Transfer the dissolution medium to the flask
solution to 41 °C while stirring, and filter the of the dissolution apparatus. During the trans-
solution under vacuum using the filter paper fer, a special care should be taken not to trap
of the pore size of 0.45 µm. During the filtra- air bubbles in the medium. Allow the dissolu-
tion procedure, continue stirring the filtrate. tion medium to reach an equilibrium tempera-
After the filtration procedure, cap the flask ture of 37 °C (it is not recommended that ro-
and continue the stirring for 5 additional tate the paddle to facilitate the medium to
minutes under vacuum. achieve the equilibrium).
c. Transfer the dissolution medium to the flask d. When the temperature of the test medium
of the dissolution apparatus. During the trans- reaches at 37 °C, initiate the dissolution test at
fer, a special care should be taken not to trap the rotational speed of 50 rpm.
air bubbles in the medium. Allow the dissolu- e. After 30 minutes of the initiation of the test,
tion medium to reach an equilibrium tempera- sample a portion of the test medium, filter and
ture of 37 °C (it is not recommended that ro- determine the absorbance at 242 nm and cal-
tate the paddle to facilitate the medium to culate the rate of dissolution (%) of salicylic
achieve the equilibrium). acid. If analytically necessary, the filtrate may
d. When the temperature of the test medium be diluted with the dissolution medium.
reaches at 37 °C, initiate the dissolution test f. If dissolution rate(%) is within the next table,
using the rotational speed of 100 rpm. it meets the requirements of the Suitability
e. After 30 minutes of the initiation of the test, test for dissolution apparatus.
sample a portion of the test medium, filter and Note)
determine the absorbance at 296 nm and cal- Dissolve a portion of Prednisone RS in ethanol to
culate the rate of dissolution (%) of salicylic render the concentration of not more than 5%, di-
acid. If analytically necessary, the filtrate may lute the solution with water to obtain a series of
be diluted with the dissolution medium. the standard solutions of known concentration and
f. If the rate of dissolution is within the range use the standard solutions for the analysis. In case
specified in the table below, the dissolution where the test solution is filtered, test for the po-
apparatus is regarded to be suitable. tential adsorption of prednisone to the filter medi-
Note) um.
Dissolve a portion of Salicylic acid RS in ethanol
to render the concentration of not more than 1%, Table 2. Dissolution specification of the prednisone
dilute the solution with the dissolution medium to standard tablet (lot number N)
obtain a series of the standard solutions of known Type of dissolution ap- Rate of dissolution at 30
KP X 1835

paratus minutes of the dissolu- Tablet Clonazepam


tion test (%) Tablet Ergotamine tartrate
Apparatus 1 (the rotary 54-78 Capsule Etoposide
basket method) Capsule Isotretinoin
Apparatus 2 (the paddle 28-54 Tablet Meprobamate
method)
1. Deaeration method I
The values of rate of dissolution indicated tables 1 and - While stirring, heat the test medium to the
2 do not represent the average rate of dissolution but temperature of 41 °C and filter the medium
represents the rate of dissolution for individual 6 or 12 under a reduced pressure with the filter paper
dissolution testing apparatuses. In addition, the specific having the pore size of 0.45 µm. During the
value for the rate of dissolution changes with the lot filtration procedure, continue the stirring of
number of the standard tablets as indicated in table 3 the filtrate with a magnetic stirrer.
below. In addition, if the dissolution testing is solely - After the filtration procedure, continue the
conducted by, for example the paddle method, then the stirring for 5 additional minutes under vacuum
suitability test may be conducted for the paddle method (sonication may be used instead of the stir-
only. ring).
- Transfer the test medium to the dissolution
Table 3. Specification of suitability test for dissolution apparatus to render the temperature of 37 °C.
apparatus using the prednisone standard tablet (lot For the case of poorly soluble drugs, the test
number J) and the salicylic acid standard tablet (lot medium may contain surfactant such as
number K) polysorbate 80 or sodium lauryl sulfate. In
Calibrator Rate of dissolution at 30 minutes of the this case, foam may be formed during the
dissolution test (%) deaeration procedure, especially in the vacu-
Apparatus 1 (the Apparatus 2 (the um filtration step. Therefore, it is recom-
rotary basket paddle method) mended that the medium, in the absence of
method) surfactant, is first deaerated and, later, added
50 rpm 100 rpm 50 rpm 100 rpm with surfactant for the dissolution testing of
Predni- 6-23 43-63 46-59 58-69 poorly soluble drugs or sustained release dos-
sone age forms.
Salicylic 14-21 23-29 13-22 16-27
acid 2. Deaeration method II
The other typical method for the deaeration of the
Appendix II test medium is helium sparging. The main compo-
nent of air dissolved in the test medium is oxygen
Deaeration of test medium and nitrogen. Helium gas, which is more inert and
Deaeration of the test medium is not always necessary less water soluble than these gases, may be used to
for all drug products for the dissolution testing. In case purge oxygen and nitrogen from the test medium.
where the identical result is obtained for the test with In general, it is sufficient that 50 mL/min of the
the deaerated dissolution medium and for that with the purging rate is sufficient for the initial 10-30
non-deaerated dissolution medium, then the deaeration minutes and then, later, the rate may be reduced to
procedure is not necessary. However, under certain 5-10 mL/min. The deaeration by helium generally
cases, the air dissolved in the dissolution medium may takes 20-40 minutes for a completion. In fact,
participate in a chemical reaction with drug substance, when helium sparing deaeration is used, it is ex-
and air bubbles trapped in the medium may affect the pected that the relative standard deviation of the
dissolution of the drug. For example, for the case of rate of dissolution is lowered with the temperature
captoril, the air dissolved in the test medium may facil- equilibration being achieved sooner. Other
itate the oxidation of captopril thereby creating a stabil- deaeration method, involving a sonication of test
ity problem. In addition, a significant amount of air medium containing surfactant in a reduced pres-
bubbles trapped in the rotary basket generally decreas- sure, may also be used.
es significantly the rate of dissolution of drugs. There-
fore, in these cases, the air dissolved in the test medium Appendix III
has to be eliminated before the initiation of the test.
Table 4 lists typical drug products in which the Sinkers used in dissolution testing
deaeration is required for the dissolution test. When the dissolution is tested using the paddle method,
certain dosage forms, for example using capsules, do
Table 4. List of drug products in which deaeration is not settle down at the bottom of the flask but float to
required for dissolution testing the surface of the dissolution solution. In this case,
Dosage form Drug substance capsules may be placed in a sinker made of wire and
Tablet Captopril the dissolution is tested. In most cases, sinkers are
1836 General Information

made of wire helix and various products, but including - Monochromator compartment: a monochromator
sinker and prolong sinker, may be used as specified in such as diffraction grating, filter, or interference filter
the Dissolution Test. is used.
- Detector compartment: it consists of a detector and
Types of Specification a signal processing system.
sinker - Display unit: it consists of display, recorder, etc.
Basket type sinker (2.6 in radius × and mathematical pretreatment on a spectrum is possi-
1.7 cm) ble.

6 helix sinker (typically 2.5 × 1 cm) 2. Measurement


Proceed as one of the following methods:

4 helix sinker (typically 2.5 × 1 cm) 1) Transmittance


Usually, this method is applied to liquids, diluted or
undiluted, and to solids in solution. The sample is
Sinker for controlled release formu-
placed in an NIR transparent cell with a proper
lation (3.8 × 1 cm; the size of the
pathlength (usually between 0.5 mm and 4 mm) or a
clamp of 5 mm × 7 mm)
fiber optic probe is inserted into the sample. When
3 extended type sinker from Vankel
recording the spectrum of a liquid sample, any spectral
Sinker specified in the Dissolution disturbances such as temperature dependent perturba-
Test (2.5-2.6 × 1.2 cm) tions should be taken into consideration, and in all cas-
es, compensation for background interferences should
be made. For example, a reference scan of air (for liq-
uids) or solvent (for solutions) may be subtracted from
the sample spectrum.
5. Guideline to Pharmaceutical 2) Diffused Reflectance
Usually, this method is applied to solids. Sample are
Quality Control Using Near analyzed using a proper apparatus. When a fiberopric
Infrared (NIR) Spectroscopy probe is immersed into the sample to acquire spectrum,
a special care should be taken to get a reproducible
I. Near Infrared Spectroscopy spectrum for each measurement. In all cases, compen-
sation for background interferences should be made.
Near infrared spectroscopy is a method very useful For example, a reference scan of an internal or external
method for identifying organic compounds by measur- reflectance reference should be subtracted from the
ing the transmittance or reflectance in a certain wave- sample spectrum. When the measurement is made, the
number or wavelength range, when the near infrared particle size, the state of hydration or of sovation
light passes through the sample. should also be taken into consideration.
The range of near infrared is between 750 nm and 3) Transflectance
2500 nm. Even though the intensity of signal is weaker Usually, this method is applied to liquids, diluted or
than that in the mid-infrared region, the appearance of undiluted, to solids in solution or in suspension. The
overtones and combinations of the fundamental vibra- measurement is made with the sample in a cell with a
tions of C-H, N-H, O-H, and S-H groups allows identi- proper diffuse reflector, made of either metal or of an
fication of materials, qualitative analysis and quantita- inert material (for example, titanium oxide), not exhib-
tive analysis. iting a spectrum in the near infrared range and intro-
Spectrum is recorded as a graph with x-axis of duced behind the sample. The reflector should show
wavenumbers (or wavelength) and y-axis of transmit- absorbance within the quantitative range.
tance or absorbance, as is in mid-infrared region. When
a spectrum acquired with a sample, the spectrum can II. Standards for Validation
be influenced by particle size, polymorphism, residual
solvents, or humidity and these influences can be re- This guideline is based on the contents of the ICH
moved by applying a proper mathematical pretreatment. general guideline to the analytical methods and is
adapted to allow the acquisition of comprehensive and
1. Instrument highly reliable information in applying analytical
Usually, an instrument consists of a light source methods using near infrared radiation, by examining
compartment, a monochromator compartment, a detec- mainly specificity, linearity, range, accuracy and preci-
tor compartment and a display unit. sion, which should be considered in using the near in-
- Light source compartment: a light source providing frared spectroscopy.
a whole range or a part of the range of 750 nm to 2500
nm (13333 cm-1 to 4000 cm-1), or a laser providing 1. Specificity
monochromic light is used.
KP X 1837

Specificity is an ability to analyze selectively the nal-to-noise ratio is lowered by 2 times by the first-
analyte, despite of probable existence of interfering order derivative operation and by 4 times by the se-
substances. Generally, a tablet contains lots of sub- cond-order derivative operation. Nonetheless, the sta-
stances in addition to the active ingredients. For exam- bility of recent instruments is excellent and this effect
ple, in the Ambroxol Tablet, the content of the princi- is not a big problem.
pal ingredient, Ambroxol, is 12.5% as 30 mg out of For the construction of calibration model, the use of
210 mg of total mass, and the remaining part, 87.5% of more than one wavelength for the analysis can remove
the total mass may interfere with the analysis of the effect caused by the substances other than the ac-
Ambroxol. These interfering substances are impurities, tive ingredients. An example of chemometric methods
degredation products, water, residual solvents, etc. and is multiple linear regression, which uses more than one
a major part of exceipients. The influence of these sub- wavelength. On the contrary, the methods which use
stances can be reduced by applying chemometric ana- information of a whole range or a certain range of
lytical methods such as the principal component analy- wavelengths are principal component regression, par-
sis. The specificity can be proved by the following tial least squres regression, and so on.
methods. The analysis of the principal components with con-
First, acquire spectra of the active ingredients and all tents of less than 5% of the tablet is expected to be
of other related substances. If the characteristic wave- problematic. Any method for increasing the specificity
lengths of the active ingredients do not overlap with by minimizing or removing the interference comprising
the wavelengths of peaks from other substances when 95% of the tablet may be studied further.
the original spectra without any pretreatment are exam-
ined, the method can be the easiest one to get the speci- 2. Linearity
ficity in analyzing the active ingredients. But when this The linearity of analytical process means the ability
kind of trend is not found with the original untreated to get the result directly propotional to the concentra-
spectra, mathematical pretreatment methods or tion (amount) of analyte in the sample. The linear re-
chemometric resolution algorithms can be applied to gression analysis is performed in the range of concen-
get the specificity by removing the interferences math- tration predictable by the calibration curve of near in-
ematically. frared spectroscopy. Usually, while the calibration
Second, by adding (fortifying the concentration) curve is obtained from the ratios of peak area in the
probable interfering substances to a mixture before range of 0% to 150% in the methods of liquid chroma-
tableting or powdered tablet, the interfering effect on tography, the result of near infrared spectroscopy gives
the analysis is identified. In other words, it should be the linear relationship between a set of reference values
examined whether the wavelength showing variation in the narrower range of 90% to 110% found by the
by the concentration of the additives overlaps with the conventional methods and a set of estimated values
characteristic wavelengths of the active ingredients. found by near infrared spectroscopy. This linear rela-
Third, in many cases, the contents of the principal tionship is evaluated by suitable statistical methods
ingredient are in the range of not less than 95% and not such as calculating the regression line by the least
more than 105% of the labeled amounts. When the squares method. Data such as correlation coefficient of
linear relationship between the actual values and the the regression line, y-intercept, slope and sum of squres
estimated values by near infrared spectroscopy of the of residuals are essential items of the documentation
concentrations outside the allowed range of the labeled for analyzing the regression data.
amounts such as 90% and 110% can be obtained, this
can be the good method to confirm the specificity. 3. Range
Fourth, the most reliable method approaching the Range is the interval between the maximum and the
specificity is the identification and the qualification of minimum of concentrations used for measuring lineari-
substances. The identification of substances is the ty, precision and accuracy during the analytical process.
comparison of the spectra acquired with the test sam- The range of analysis is set to 2 times the allowed
ples, with the spectra acquired with the (powdered) range, for there are lots of difficulties in analyzing tab-
library test samples having variations caused during the lets nondestructive and in collecting test samples for
production process. The qualification of substances is calibration. For example, the allowed range of contents
the comparison of the mean spectrum of the test sam- of the principal ingredients is given to be 5%, consid-
ples, with the library spectra. By these procedures, the ering the degradation of the principal ingredients dur-
placebos can be identified. ing the effective period of use and the error of analyti-
The second derivative operation to spectral data has cal methods. In this case, the minimum range in ana-
several advantages for the analysis. The differentiation lyzing the principal ingredients is 10% of the labeled
between the peaks of the active ingredients and proba- amount. The more accurate calibration be made if the
ble interfering substances can be achieved, and the tablets are collected so that the range is widen to be
background perturbation can be removed. In addition between 80% and 120% of the labeled amount. In re-
to this, peaks can be sharpened, and any variation ality, the range suitable for the purpose of analysis is
caused by the physical states of tablets can be removed, preferred, considering the ranges of 80% to 120% for
resulting in the increased resolution. Usually, the sig- the Assay of the drug substances or the preparations,
1838 General Information

70% to 130% for the Uniformity of Dosage Units and performing the whole procedure 6 times in a short pe-
20% of the range of specification for the Dissolution riod time, as long as there is no thermal degradation of
Test. the samples with concentration equivalent to 100% of
the tested concentration. The sample homogeneity and
4. Accuracy the surface homogeneity should be taken into more
The accuracy of analytical process is the measure consideration especially when the contents of the prin-
expressing the similarity between the values found by cipal ingredients are low. In this case, the transmittance
the conventional analytical method (with proven accu- is better than the reflectance, for the area of transmit-
racy) and the estimated values found by near infrared tance is wider than that of reflectance. Meanwhile, the
spectroscopy. The accuracy should be proved in the transmittance may introduce more noise when the tab-
whole range specified by the analytical method. The lets are thick and the light intensity reaching the detec-
accuracy is a parameter evaluated only after establish- tor is low.
ing precision, linearity and specificity. The representative factors of variation which need
The accuracy can be evaluated by at least 9 meas- examination in within-laboratory reproducibility are
urements in the specified range. For example, the re- the date of experiment, the experimenter, and the in-
sults measured by manipulating 3 levels of concentra- strument used and so on. The inter-laboratory repro-
tion and 3 repeatitions of each concentration level. ducibility is evaluated when the analytical method is
Three minimum levels of concentrations are selected as needed to be standardized for the collaborative experi-
the mean value and both of the maximum and the min- ments between laboratories. If the inter-laboratory re-
imum. The accuracy is expressed as the recovery, when producibility is proven, the within-laboratory reproduc-
the samples spiked with a known level of the analyte ibility is not needed to be validated, but usually, the
are quantitated, or it is examined by comparison be- within-laboratory reproducibility is mainly evaluated,
tween the reference value and the mean value of meas- for the inter-laboratory reproducibility is hard to be
urements in the confidence interval, when the compari- achieved.
son with the true value or the certified or agreed value
is made. In the quantitative analysis by near infrared III. Qualification of NIR Spectrophotometers
spectroscopy, the accuracy is expressed as the standard
error of prediction found with the validation set. The purpose of qualification of NIR spectrophotom-
The calibration set should consist of the samples eters is to verify the suitability of the instrument for the
with the concentrations of the active ingredients pro- intended use by comparing with the specification of the
duced in the batch process and it should include the instrument. The qualification procedure includes De-
whole range of the sample. The selected samples sign Qualification (DQ), Installation Qualification (IQ),
should show the uniform distribution of probability, Operational Qualification (OQ) and Performance Qual-
not the normal distribution. The validation set should ification (PQ).
consist of the samples with the same composition of
the active ingredients as in the calibration set. 1. Design Qualification (DQ)
The set of samples for the parallel test are selected The design qualification provides the evidence
from the independent production batch and the analysis whether the instrument is properly designed to be oper-
is validated by the time. Therefore, the accuracy of the ational for the intended use. The various influencing
analysis can be examined using the set completely in- factors of the instrument are tested to verify whether
dependent from the calibration set and the validation they correspond to the specifications of the instrument.
set of samples. This test is performed once a month At least, it should be verified whether the factors corre-
with independent set of samples after development of spond the manufacturer’s specifications of the instru-
the calibration model. ment.

5. Precision 2. Installation Qualification (IQ)


The precision of analytical process means the simi- The installation qualification verifies whether the in-
larity among the results obtained by a series of repeat- strument is installed according to the design and the
ed analysis with the same sample under the specified expressed specifications. Documentation of the serial
analytical conditions. The items of precision are re- number of the hardware, the version of the software
peatability, within-laboratory reproducibility and inter- and so on is included in the verification. And the envi-
laboratory reproducibility. The precision is tested with ronment and the facilities of instrument installation are
homogeneous and authentic samples. When the homo- are verified and the status of assembly, electric power
geneous and authentic samples are not easily available, of the instrument and so on are also verified.
the test can be done with artificially synthesized sam-
ples or solutions. The precision is expressed as the rela- 3. Operational Qualification (OQ)
tive standard deviation or the coefficient of variation The operational qualification verifies the instrument
calculated from a series of measurements. once again by the methods used to select the spectro-
Repeatability is set to not more than 1.0% as the rel- photometer, such as wavelength accuracy and precision,
ative standard deviation (or CV) by measuring and linearity of the monochromator and noise.
KP X 1839

The feasibility test is also needed for the selection of


4. Performance Qualification (PQ) algorithm for the calibration. When the principal peaks
The performance qualification verifies whether the of the active ingredients do not overlap with those of
instrument continues to be operational. Parts of the OQ the excipients, the multiple linear regression can be
items are needed to be applied to verify the instrument used. When there is a significant interference, the
before the use or periodically (at least once every 6 pricipal component regression or the partial least
months). The PQ should be performed when the squares regression is more effective. Items that are
maintenance of the instrument is done or the lamp is needed to be checked are whether to use transmittance
replaced. or reflectance, the use of fiber-optics, whether there are
reproducible results by repeated measurements of the
IV. Quantitative Analysis sample, whether there is robustness in the measurement
of the sample, and so on.
In the pharmaceutical industries, the quality of
pharmaceuticals are assessed by the repeatition of ex- 2. Reference data
periments for several times with the drug substances The quantitative method by NIR spectroscopy is
and the final pharmaceutical preparations. Nonetheless, based on the comparison of the reference data with
the conventional analytical methods for the quality acquired spectral data. Therefore, if the reference data
control cannot be applied to all of the produced phar- are not correct, the estimated values by NIR spectros-
maceuticals, for those methods are usually destructive. copy cannot be correct either. The reference data may
Therefore, the weakness that only the representative be obtained by the preparation of real reference stand-
samples are managed can be a critical obstacle in the ards or the application of reference analytical methods,
matter of safety. Several principles of the analytical i.e. conventional analytical methods used for the evalu-
process are needed for the optimal maintenance pro- ation, such as gravimetry, chromatography, spectros-
cess. The accuracy and the precision are necessary and copy, and so on, of which accuracy is proven. The lin-
the sample preparation process is not required or is earity, accuracy and precision of the reference analyti-
minimal. Moreover, the simultaneous analysis of many cal methods should be verified. The environmental
analytes should be possible and the rapid adjustment error can be minimized by concurrent use of the NIR
during the production process should be possible. The spectroscopy and the reference analytical method.
near infrared spectroscopy is one of the methods satis- Three repeated measurements with the same sample by
fying these principles. The near infrared spectroscopy the reference analytical method are made to get the
allows non-destructive, real-time total inspection of reference data
tablets during the production process. The non-
destructive quantitative analysis of the tablets is to be 3. Selection of the samples
mentioned among the quantitative analyses of various i) Calibration Test Set
preparations and the general procedure is as follows: The calibration test set is the standard for the quanti-
fication of the NIR spectral responses against the refer-
• Feasibility test ence data. The sample should be selected carefully so
• Reference data that the calibration test set includes the whole range of
• Selection of the samples possible concentrations of the analytes, and there is
• Acquisition of sample spectra robustness over the variation in the concentration of the
• Construction of calibration model excipients, and the maximum variation of the analysis
• Validation of calibration model is included. As other analytical method, interpolation
• Validation of model performance within the range of the calibration test set is possible
with the NIR spectroscopy, but extrapolation is not
1. Feasibility test possible.
The feasibility test is a procedure to check whether a
drug material or a pharmaceutical product can be ana- ii) Validation Test Set
lyzed quantitatively by NIR spectroscopy. The first The construction of validation test set is the first step
step is to observe the spectral response at the used level of the validation. The validation test set is used to op-
of concentration of the analyte. After acquiring and timize the model and the concentration used should be
examining spectra of all the ingredients of a tablet, if within the range of the calibration test set. If any varia-
there are no significant differences among the ingredi- tion outside the calibration test set is included, the cor-
ents, the second derivatives may be used to identify rect validation values can be obtained.
characteristic peaks of each ingredient from the
derivatized spectra. In addition to this, when the spec- Partitioning of the calibration test set and the valida-
tra are acquired at the different level of concentrations tion test set is done as follows. With concurrent exper-
of samples, any spectral changes caused by the iments, the calibration test set and the validation test
concentrationn are checked with the second derivatives set have the same instrumental error and the environ-
of those spectra. mental error.
1840 General Information

The test set is selected manually or by software the concentration range is wider (80% to 120%), for
method so that the composition of the analytes is uni- the amount of the active ingredients can be varied
formly distributed. from the first stage of the sample preparation.
The test set can be selected on the basis of the spec- Fourth, the active ingredients and the excipients are
tral variation by software method. added to the powdered tablets and then the mixture
Whichever method is used, the care should be taken is tableted. But in this case, the total mass of the tab-
to have uniform distribution by the concentration range. let is different from that of the products and it may
When the distribution is not uniform (for example, fail in terms of specificity.
concentrations at the center are used), there should be a Fifth, the model comprising all of the variation fac-
proper explanation. tor of the production process by using the global cal-
ibration set, which includes all of the existing varia-
The sample size measurable by NIR spectroscopy is tion factors to make the calibration models. This
significantly smaller than those by the conventional method consists of 4 steps: laboratory composite
method. This is not because of the sample measure- (prepared to have active ingredients with the concen-
ment compartment but because of the sample area il- tration level of 15% of the labeled amount), prepa-
luminated by the near infrared light. The infrared ration of granules, tableting of core tablet, and coat-
measurement can detect the inhomogeneity even with ing the tablets.
the mass of microgram level and a proper measurement
method is needed to accommodate this variation. This 4. Acquisition of sample spectra
property can be an advantage useful for testing the The transmittance is applied to liquids, diluted or
Uniformity of Dosage Units, but in many applications, undiluted and solids in the solution. The cells with
averaging the signal from a larger area is needed. transmittance pathlength of 0.5 to 4 mm or dip-probes
Therefore, to get the averaged information, the sample are used and the compensation for background inter-
is moved transversely or rotated during the data acqui- ferences should be made. On the contrary, the reflec-
sition. tance and the diffused reflectance are applied mainly to
Among the various preparations, in cases of the tab- solids and the test is done to the sample placed in a
lets, there are two cases: the case of using the products proper apparatus. When the fiber-optics are used, re-
as they are and the case of varying ratios of the active producible spectra can be acquired by fixing the sam-
ingredients to the excipients. ple properly. As in the transmittance, compensation for
background interferences should be made.
(1) The case of using the products as they are In cases of solid particles, the physical differences
First, this can be applied only when the transmit- such as particle size, shape, compressibility, etc. can be
tance follows the Beer-Lambert’s law and it also ap- influential, so the particles are manipulated to be as
plied to other tablets of the same composition with small and uniform in size as possible. Moreover, to
different masses. make the effect of the absorbed water and the residual
Second, this can be applied when the samples are solvent constant, the samples are dried for a certain
collected from the normal production and each tablet length of time to have the same condition of the ab-
has the concentration range needed for the usage. sorbed water and the residual solvent of the samples.
When the samples are analyzed by the NIR spectrosco-
(2) The case of varying ratios of the active ingredi- py, the NIR spectra are influenced a lot by the ab-
ents to the excipients sorbed water and the temperature, so it is desirable to
First, the calibration test set is obtained by mixing construct the environment of constant humidity and
the products and the concentration expansion sam- constant temperature so that the experiment can be
ples. Usually, the products lie between 95% and 105% done in the same environment. Moreover, the differ-
of the labeled amount, so to expand this range, tab- ence in the polymorphism of the sample or in the de-
lets with expanded concentration range are synthe- gree of crystalization also can be influential to the
sized in the laboratory. This concentration expansion quantitative analysis, so the case should be taken.
samples have different ratios of the active ingredi- After selecting a proper sample measurement meth-
ents to the excipients and the ranges of between 90% od among various sample measurement apparatus by
and 110% of the labeled amount or between 85% considering the property, the shape, and the size of the
and 115% of the labeled amount are used. sample, the precision of the spectra acquired by repeat-
Second, the concentration is adjusted by adding the ed measurement is evaluated. When the measurement
active ingredients and the excipients to the powdered is repeated 6 times, it meets the requirement if the
products. If the active ingredients are low compared maximum value of the relative standard deviations of
to the total mass of the tablet, the better calibration whole range of wavelengths is not more than 1.0%.
results can be acquired by adding the active ingredi-
ents to the powdered tablets. 5. Construction of calibration model
Third, the concentration of the active ingredients is For the development of a calibration model, a proper
varied using the sample synthesized in the laboratory. mathematical preptreatment is needed to be done, if
This is similar to the use of the powdered tablet, but necessary. The pretreatment procedure is an important
KP X 1841

step in chemometric analysis of the NIR data. The pre- converting the amount (mg) of the analyte to 100 is not
treatment is defined as a procedure in which the spec- exceeding the error range of the conventional method
tral shape is enhanced by mathematical treatment of the of analysis, the use of the model is recommended.
NIR data or unwanted variation is removed prior to the
development of a calibration model. A proper pre- 7. Validation of model performance
treatment method can be selected by testing the spec- Not just to evaluate simply the model, the perfor-
tral data prior to the data modeling. For example, vari- mance of the model is needed to be validated periodi-
ous methods of pretreatment are used in parallel and cally to use that model continuously in the industrial
evaluated and the best one is selected among them. fields. The methods for validating model performance
There are various methods of pretreatment. For ex- can be classified into two methods, which are the
ample, they are normalization, smoothing, baseline method of using the check samples and the method of
correction, derivatives, mean centering, variance scal- comparing with the reference analytical method. First,
ing, autoscaling and so on. to use the check samples, the check samples should be
After selecting a proper method of pretreatment, re- stable over time and the short-term and long-term accu-
gression analysis on the NIR data is done by applying racy of the model should be evaluated with these sam-
various quantitative algorithms. The calibration is a ples. Second, to use the method of comparing with the
procedure by which a mathematical model is con- reference analytical method, the data by the NIR spec-
structed between the instrumental responses and the troscopy and the reference data are acquired periodical-
properties of the sample (usually concentration). As ly for n months (n=1,2,3, ) over n batches and a
mentioned previously, the major algorithms for calibra- paired t-test with the confidence level of 95% is per-
tion are multiple linear regression (MLR), principal formed. When there is significant difference, the quan-
component regression (PCR), partial least squares re- titative model should be reconstructed.
gression (PLSR) and so on. The prediction is a proce-
dure of predicting the properties of the samples from * Refer to the Appendix II for the validation of the
the instrumental signals using the developed model. In model.
a broad sense, there are two different approaches to * Refer to the Appendix III for the usual quantitative
develop the calibration model and they are the analysis and the maintenance of the quantitative model.
univariate analysis and the multivariate analysis. The
univariate analysis is the most commonly used method V. Qualitative Analysis
in the conventional analysis, which relates the single
signal of the analytical instrument is related to the con- The near infrared spectroscopy can be used for the
centration of the single ingredient. This method is not identification of substances and the qualitative analysis.
commonly used in the NIR spectroscopy. In the NIR
spectroscopy, many signals from the analytical instru- Identification : It is used when the chemical identifi-
ment are related to the various properties of the sam- cation of the substance is needed.
ples and the calibration model is constructed by the Qualitative analysis : After chemical identification
multivariate analytical methods. of the substance, the suitability of the sample to the
model of substances is measured. The model is devel-
* For constructing a quantitative model, refer to the oped from the samples representing various infor-
Appendix I. The Appendix I explains mainly MLR, mation on the variation and it includes water, particle
PCR and PLSR, which are used generally in the NIR size, solvent and other chemical and physical
spectroscopy. informations.
Both of the identification and the qualitative analysis
6. Validation of calibration model enable the discrimination of substances in the library.
The quantitative model can be validated internally or Typical qualitative application of the NIR spectroscopy
externally. Independent validation test set is used to get consists of the following procedures.
the information on the predictability of the developed
model. The accuracy and the precision of the NIR • Feasibility test
spectroscopy is compared with those of the reference • Selection of the samples
analytical method. Standard error of calibration (SEC) • Acquisition of sample spectra
and standard error of prediction (SEP) are used to • Construction of library
evaluate the quality of the quantitative model. A con- • Validation of library
ventional factor of model evaluation, the correlation • Routine use
coefficient of regression is also used in the NIR spec- • Maintenance of library
troscopy, but it is not as important as in the conven-
tional univariate analysis. 1. Feasibility test
When the content of a tablet is analyzed, the ratio (%) The feasibility test is performed as a first step, prior
of the amount of analyte to the mass of the tablet is to the development of model. For example, optimal
used as the value of concentration. When the calculated method of sample measurement, amount of the sample,
SEP value derived as the relative error obtained by the minimum number of scans for effective analysis are
1842 General Information

examined. A prior knowledge on the organization of sorbed water and the temperature, so it is desirable to
samples in the library and the molecular structure is construct the environment of constant humidity and
useful in the analysis with the NIR spectroscopy. And constant temperature so that the experiment can be
when the spectra of the representative samples are ac- done in the same environment. Moreover, the differ-
quired, it is checked whether the second derivative ence in the polymorphism of the sample or in the de-
spectra of the samples are different from each other. gree of crystalization also can be influential to the
quantitative analysis, so the case should be taken.
2. Selection of the samples After selecting a proper sample measurement meth-
The selection of the samples is an important proce- od among various sample measurement apparatus by
dure for the successful qualitative analysis. The sample considering the property, the shape, and the size of the
set for developing the library and the independent set sample, the precision of the spectra acquired by repeat-
for the purpose of validation are needed. All of the ed measurement is evaluated. When the measurement
samples used for building and validating the library are is repeated 6 times, it meets the requirement if the
needed to be authenticated. The level of authentication maximum value of the relative standard deviations of
of the sample is varied by the usage and the database whole range of wavelengths is not more than 1.0%.
consists of the samples showing various source of vari-
ation. 4. Construction of Library
The samples from the different batches are collected, The following procedures are used to develop the li-
reflecting the changes in ingredients, suppliers, pro- brary.
cesses, and storage conditions over a certain period of
time. If the chemical and physical stability is proven • Definition of the purpose of developing the library
over the storage period, the samples can be collected • Selection of the sample set for developing the li-
regardless of the storage status. brary
The number of batches to be collected is dependent • Data display
on the complexity of analysis and the substances to be • Selection of the sample set for validating the library
analyzed should include the typical variation of the • Data pretreatment/conversion
system. The number of batches for the qualitative anal- • Construction of the library
ysis is greater than that for the identification. There are • Setting the threshold
many kinds of the sample measurement apparatus, such
as cups, vials, fiber-optics and custom-made apparatus. 1) Definition of the purpose of developing the library
The selection of the apparatus is dependent on the us- Prior to the development of the library, it is im-
er’s needs and the validation of the apparatus is defined portant to set the effective range of the library accord-
in the stage of the design qualification (DQ). The sam- ing to the intended use. This applies to the identifica-
ple measurement apparatus is the source of latent varia- tion and the qualitative analysis of the substances and it
tions during the measurement. Therefore, it should be includes checking the chemical similarity of the groups
validated continuously in terms of reproducibility, if to be analyzed and the number of groups.
possible.
2) Selection of the sample set for developing the li-
3. Acquisition of Sample Spectra brary
The transmittance is applied to liquids, diluted or For the development of the library, the variation
undiluted and solids in the solution. The cells with caused by the following factors should be considered.
transmittance pathlength of 0.5 to 4 mm or dip-probes The factors, especially important for the library for the
are used and the compensation for background inter- qualitative analysis are as follows:
ferences should be made. On the contrary, the reflec-
tance and the diffused reflectance are applied mainly to • Moisture
solids and the test is done to the sample placed in a • Particle size
proper apparatus. When the fiber-optics are used, re- • Residual solvents
producible spectra can be acquired by fixing the sam- • Degradation products
ple properly. As in the transmittance, compensation for • Compositional change of formulated product
background interferences should be made. • Other chemical/physical properties
In cases of solid particles, the physical differences • Time
such as particle size, shape, compressibility, etc. can be • Alternative sources of material
influential, so the particles are manipulated to be as • Retained samples
small and uniform in size as possible. Moreover, to • Temperature
make the effect of the absorbed water and the residual • Operator
solvent constant, the samples are dried for a certain • Presentation, e.g. insertion of tube
length of time to have the same condition of the ab- • Between-instrument variation
sorbed water and the residual solvent of the samples. • Others
When the samples are analyzed by the NIR spectrosco-
py, the NIR spectra are influenced a lot by the ab-
KP X 1843

The range of consideration over these factors is de- unwanted effects or in bringing a small but important
pendent on the intended use and the needed ability of difference into relief.
classification. Like any other analytical techniques, the NIR spec-
troscopy cannot differentiate all of the substance
3) Data display groups, especially very similar groups. In this case, two
The data display checks visually the sample showing groups are combined to one group or any other control
strange spectral features and identifies outliers. If pos- method is used to perform the identification and quali-
sible, outliers should be identified and if there is rea- tative analysis.
sonable analytical explanation, they can be removed There are many algorithms such as correlation, soft
and in that case, the clear reason should be documented. independent modeling of class analogy (SIMCA),
Mahalanobis distance, SMV and so on. The user se-
4) Selection of the sample set for validating the li- lects a proper algorithm with the consideration of the
brary effective range of the library. When the discrimination
There can be a situation, in which the representative of the substances is each, it is recommended to use the
samples are needed to be selected from the large group. simplest algorithm. For example, when the purpose is
In the simple situation, they can be selected manually, only the identification of the substance, the physical
but in more complex situation, they should be selected factors are not needed to be considered, so the second
after the determination of substance groups using a derivatives and the wavelength correlation algorithm
proper selection tool (for example, principal compo- can be selected.
nent analysis, cluster analysis, etc.).
The number of samples for each of the substance 7) Setting the threshold
groups is dependent on the qualitative algorithm and The internal validation is done with the value set by
the state of the system (how accurately the boundaries the software itself or recommended by the manufactur-
of groups needed to be set). er. The threshold of the library can be changed when
the library is validated internally or the sample is eval-
5) Data pretreatment/conversion uated externally.
Data are needed to be pretreated mathematically to
simplify the spectra more. For example, the algorithms 5. Validation of Library
of derivatives and scatter correction can remove the The purpose of validation of an analytical procedure
background variation caused by the physical difference. is to check whether the analysis is suitable for the in-
The original untreated spectra are used when the in- tended use. Upon this purpose, the factors influencing
formation due to the physical status is important. the needed validation should be determined.
Mathematical transformation of spectra data can be
an artifact and can cause loss of important information, 1) Internal validation
so this should be taken into consideration. The algo- The performance of the library should be evaluated
rithms of data pretreatment and conversion should be in the course of constructing any kind of spectral data-
understood correctly and the theoretical basis always base. This is based on the samples constructing the
should be provided whenever any conversion is per- library (whether these samples can be differenctiated).
formed. The internal validation is done by the software and the
detailed procedure may be different by the used soft-
6) Construction of the library ware, but the basic procedure is as follows:
The structure of the library is dependent on the limi-
tation of the software and the user’s needs. In the sim- • The spectra used to construct the library are vali-
plest case, all of the substances are combined into one dated by a proper method (correlation or distance
library. On the contrary, it can be divided into sub- method)
libraries to secure the needed level of the specificity. • It is checked whether the distribution of the sub-
The mathematical transformation procedure may be stances in the library overlaps.
same for all of the substance groups in the principal • The cross-validation is used for the construction of
library. The transformation may be same within the the library.
sub-library but different between the sub-libraries. For
example, this situation is met when the grade of the 2) External validation
lactose is to be determined after the sample is identi- When the internal validation is successful, the per-
fied as lactose from the library of excipients. The spec- formance of the database is validated using certified
tral range can be all of the available wavelengths or can samples not used for the database development.
be limited to cetain wavelengths. The range of wave-
lengths can be limited when a certain measurement Reproducibility: This is not commonly applied
apparatus is used or unnecessary spectral information method for the identification of substances. In the re-
is needed to be removed (outside the dynamic range or producibility test for the qualitative analysis, the sub-
regions of high noise level). An analytical method of stances included in the library should be reliably dif-
partitioning the wavelength range is useful in removing
1844 General Information

ferentiated from the substances outside the library, To add new substances into the library, the test set of
using the threshold value. substances is selected to satisfy the details of sample
Robustness: This category is dependent on the appli- selection parameters and the library is evaluated again
cation and the sample selection technique and the ef- for the validation of the specificity, continuously.
fect of delicate variation in the normal operating condi-
tion of the analysis is tested. The use of the design of 3) Fixing the groups in the library
experiment can maximize the effect of the information Sometimes, the sample set is fixed in the cases as
on the analyte. The following items are considered: follows:

• The effect of environmental conditions (tempera- • Change in physical properties of the substance
ture, humidity) of the analysis • Change in supplier
• The effect of temperature on the sample • Inclusion of the wider range
• The location of sample
• The depth of probe and the degree of compres- In each case, new samples are certified by the meth-
sion/filling of the substance od other than the NIR spectroscopy, before inserting
• Other effect of the sample measurement apparatus them in the model and the library is evaluated again for
• The effect of replacement of parts (lamp) the validation of the specificity, continuously.
• The effect of pretreatment and parameters of the al-
gorithm used to construct the library (gap/segment * Refer to the Appendix 5 when there is any problem
of the derivative operation, distance threshols, etc.) in applying the library for the qualitative analysis.

6. Routine use VI. Terms Used


When the system is evaluated, it is adjusted to allow
the needed functions only. For example, the manager Calibration: a procedure of construction of a quanti-
of the NIR system or the library development expert tative model
should evaluate the software thoroughly, but general Prediction: a procedure of estimating the concentra-
users are required to evaluate only the performance of tion of unknown sample, using the constructed quanti-
the general identification of substances tative model
In the aspect of developing the library for the spec- Calibration set: a set of samples used for construct-
tral identification, the considerable part of the charac- ing the quantitative model
teristic variation within the substances are to be includ- Validation test set or validation set: a set of samples
ed. But sometimes, this variation may not be included used for validating the quantitative model
in the sample set for the development of the library. Multiplicative Scatter Correction: one of the mathe-
For example, when a test substance is analyzed to be matical methods used for the correction of background
outside the boundary of one of the models in the library, variation caused by the physical properties such as
the test substance is expressed as to be “inadequate particle size of the sample
NIR spectrum” for the NIR spectral model. In this case, Multiple Linear Regression (MLR): quantitative
a proper alternative test is performed and the test sub- model constructed with absorbance values at two or
stance is certified correctly before making decision that more wavelengths.
the substance is within the model or inserting the spec- Principal Component Regression (PCR): one of the
trum into the library. multivariate regression analysis using principal com-
ponent analysis, or factor analysis.
* Refer to the Appendix IV for the construction of Partial Least Squares Regression (PLSR): one of the
the library for the qualitative analysis. multivariate regression analysis using principal com-
ponent analysis to which the concentration information
7. Maintenance of library of the samples are included.
1) Removal of existing substances Soft Independent Modeling by Class Analogy
In normal situations, the removal of substances from (SIMCA): a pattern recognition algorithm used for the
the library is not recommended, usually, but when the identification and the qualitative analysis of the sam-
selection of the samples is proven to be wrong, the ples.
removal from the library is made and the library should NIST SRM: National Institute of Standards and
be evaluated once again. Technology Standard Reference Material

2) Addition of new substances


KP X 1845

Appendix I. Selection of an algorithm for the construction of quantitative model.


1846 General Information

Appendix II. Validation and Evaluation of a Quantitative Model


KP X 1847

Appendix III. Routine procedure of quantitative analysis and Maintenance of the quantitative model.
1848 General Information

Appendix IV. Construction of library needed for qualitative analysis


KP X 1849

Appendix V. In case of failure of the library for qualitative analysis


1850 General Information

Therefore, testing should be performed for residual


6. Guideline of Limits for solvents when production or purification processes are
Residual Solvents of known to result in the presence of such solvents. It is
only necessary to test for solvents that are used or pro-
Pharmaceuticals duced in the manufacture or purification of drug sub-
stances, excipients, or drug product. Although manu-
1. INTRODUCTION facturers may choose to test the drug product, a cumu-
lative method may be used to calculate the residual
The objective of this guideline is to recommend ac- solvent levels in the drug product from the levels in the
ceptable amounts for residual solvents in pharmaceuti- ingredients used to produce the drug product. If the
cals for the safety of the patient. The guideline recom- calculation results in a level equal to or below that rec-
mends use of less toxic solvents and describes levels ommended in this guideline, no testing of the drug
considered to be toxicologically acceptable for some product for residual solvents need be considered. If,
residual solvents. however, the calculated level is above the recommend-
‘Residual solvents’ in pharmaceuticals are defined ed level, the drug product should be tested to ascertain
here as organic volatile chemicals that are used or pro- whether the formulation process has reduced the rele-
duced in the manufacture of drug substances or excipi- vant solvent level to within the acceptable amount.
ents, or in the preparation of drug products. The sol- Drug product should also be tested if a solvent is used
vents are not completely removed by practical manu- during its manufacture.
facturing techniques. Appropriate selection of the sol- This guideline does not apply to potential new drug
vent for the synthesis of drug substance may enhance substances, excipients, or drug products used during
the yield, or determine characteristics such as crystal the clinical research stages of development, nor does it
form, purity, and solubility. Therefore, the solvent may apply to existing marketed drug products.
sometimes be a critical parameter in the synthetic pro- The guideline applies to all dosage forms and route
cess. This guideline does not address solvents deliber- of administration. Higher levels of residual solvents
ately used as excipients nor does it address solvates. may be acceptable in certain cases such as short term
However, the content of solvents in such products (30 days or less) or topical application. Justification for
should be evaluated and justified. these levels should be made on a case by case basis.
Since there is no therapeutic benefit from residual See Appendix 2 for additional background information
solvents, all residual solvents should be removed to the related to residual solvents.
extent possible to meet product specifications, good
manufacturing practices, or other quality-based re- 3. GENERAL PRINCIPLES
quirements. Drug products should contain no higher
levels of residual solvents than can be supported by 3.1 Classification of Residual Solvents by Risk
safety data. Some solvents that are known to cause Assessment
unacceptable toxicities (Class 1, Table 1) should be The term tolerable daily intake (TDI) is used by the
avoided in the production of drug substances, excipi- International Program on Chemical Safety (IPCS) to
ents, or drug products unless their use can be strongly describe exposure limits of toxic chemicals and ac-
justified in a risk-benefit assessment. Some solvents ceptable daily intake (ADI) is used by the World
associated with less severe toxicity (Class 2, Table 2) Health Organization (WHO) and other international
should be limited in order to protect patients from po- health authorities and institutions. The new term per-
tential adverse effects. Ideally, less toxic solvents mitted daily exposure (PDE) is defined in the present
(Class 3, Table 3) should be used where practical. The guideline as a ‘pharmaceutically acceptable intake of
complete list of solvents included in this guideline is residual solvents’ to avoid confusion of differing val-
given in Appendix 1. ues for ADI's of the same substance.
The lists are not exhaustive and other solvents can Residual solvents assessed in this guideline are listed
be used and later added to the lists. Recommended in Appendix 1 by common names and structures. They
limits of Class 1 and 2 solvents or classification of sol- were evaluated for their possible risk to human health
vents may change as new safety data becomes availa- and placed into one of three classes as follows:
ble. Supporting safety data in a marketing application
for a new drug product containing a new solvent may Class 1 solvents: Solvents to be avoided
be based on concepts in this guideline or the concept of Known human carcinogens or strongly sus-
qualification of impurities as expressed in the guideline pected human carcinogens, and environmental
for drug substance (Q3A, Impurities in New Drug Sub- hazards.
stances) or drug product (Q3B, Impurities in New Drug Class 2 solvents: Solvents to be limited
Products), or all three guidelines. Non-genotoxic animal carcinogens or possible
causative agents of other irreversible toxicity
2. SCOPE OF THE GUIDELINE such as neurotoxicity or teratogenecity. Sol-
vents suspected of other significant but re-
Residual solvents in drug substances, excipients, and versible toxicities.
in drug products are within the scope of this guideline.
KP X 1851

Class 3 solvents: Solvents with low toxic potential permitted daily exposure to acetonitrile is 4.1 mg/day;
Solvents with low toxic potential to man; thus, the option 1 limit is 410 ppm.
no health-based exposure limit is needed. The maximum administered daily mass of a drug
Class 3 solvents have PDEs of 50 mg or more product is 5.0 g, and the drug product contains two
per day. excipients. The composition of the drug product and
the calculated maximum content of residual acetonitrile
3.2 Methods for Establishing Exposure Limits are given in the following table.
The method used to establish permitted daily expo-
sures for residual solvents is presented in Appendix 3. Compo- Amount in Acetonitrile Daily
Summaries of the toxicity data that were used to estab- nent formulation content exposure
lish limits are published in Pharmeuropa, Vol. 9, No. 1,
Supplement, April 1997. Drug
0.3 g 800 ppm 0.24 mg
substance
3.3 Options for Setting Limits of Class 2 Solvents Excipient 1 0.9 g 400 ppm 0.36 mg
Two options are available when setting limits for
Excipient 2 3.8 g 800 ppm 3.04 mg
Class 2 solvents.
Drug
5.0 g 728 ppm 3.64 mg
Option 1: The concentration limits in ppm stated in Product
Table 2 can be used. They were calculated us-
ing equation (1) below by assuming a product Excipient 1 meets the Option 1 limit, but the drug
mass of 10 g administered daily. substance, excipient 2, and drug product do not meet
the Option 1 limit. Nevertheless, the product meets the
Concentration (ppm) = 1000 PDE (1) Option 2 limit of 4.1 mg per day and thus conforms to
dose the recommendations in this guideline.
Consider another example using acetonitrile as re-
Here, PDE is given in terms of mg/day and dose is sidual solvent. The maximum administered daily mass
given in g/day. of a drug product is 5.0 g, and the drug product con-
These limits are considered acceptable for all sub- tains two excipients. The composition of the drug
stances, excipients, or products. Therefore this option product and the calculated maximum content of residu-
may be applied if the daily dose is not known or fixed. al acetonitrile are given in the following table.
If all excipients and drug substances in a formulation
meet the limits given in Option 1, then these compo- Compo- Amount in Acetonitrile Daily
nents may be used in any proportion. No further calcu- nent formulation content exposure
lation is necessary provided the daily dose does not
Drug
exceed 10 g. Products that are administered in doses 0.3 g 800 ppm 0.24 mg
substance
greater than 10 g per day should be considered under
Option 2. Excipient 1 0.9 g 2,000 ppm 1.80 mg
Excipient 2 3.8 g 800 ppm 3.04 mg
Option 2: It is not considered necessary for each com-
Drug
ponent of the drug product to comply with the 5.0 g 1,016 ppm 5.08 mg
Product
limits given in Option 1. The PDE in terms of
mg/day as stated in Table 2 can be used with
In this example, the product meets neither the Op-
the known maximum daily dose and equation
tion 1 nor the Option 2 limit according to this summa-
(1) above to determine the concentration of re-
tion. The manufacturer could test the drug product to
sidual solvent allowed in drug product. Such
determine if the formulation process reduced the level
limits are considered acceptable provided that
of acetonitrile. If the level of acetonitrile was not re-
it has been demonstrated that the residual sol-
duced during formulation to the allowed limit, then the
vent has been reduced to the practical mini-
manufacturer of the drug product should take other
mum. The limits should be realistic in relation
steps to reduce the amount of acetonitrile in the drug
to analytical precision, manufacturing capabil-
product. If all of these steps fail to reduce the level of
ity, reasonable variation in the manufacturing
residual solvent, in exceptional cases the manufacturer
process, and the limits should reflect contem-
could provide a summary of efforts made to reduce the
porary manufacturing standards.
solvent level to meet the guideline value, and provide a
risk-benefit analysis to support allowing the product to
Option 2 may be applied by adding the amounts of a
be utilized with residual solvent at a higher level.
residual solvent present in each of the components of
the drug product. The sum of the amounts of solvent
3.4 Analytical Procedures
per day should be less than that given by the PDE.
Residual solvents are typically determined using
Consider an example of the use of Option 1 and Op-
chromatographic techniques such as gas chromatog-
tion 2 applied to acetonitrile in a drug product. The
raphy. Harmonized procedures for determining levels
1852 General Information

of residual solvents as described in the pharmacopoeias Concentration


should be used, if feasible. Otherwise, manufacturers Solvent Note
limit (ppm)
would be free to select the most appropriate validated Carcino-
analytical procedure for a particular application. If only Benzene 2
gen
Class 3 solvents are present, a non-specific method Toxic and
such as loss on drying may be used. Carbon tetrachlo- environ-
Validation of methods for residual solvents should 4
ride mental
conform to the ICH guidelines Text on Validation of hazard
Analytical Procedures and Extension of the ICH Text 1,2-Dichloroethane 5 Toxic
on Validation of Analytical Procedures . 1,1-Dichloroethene 8 Toxic
Environ-
3.5 Reporting levels of residual solvents 1,1,1-
1500 mental
Manufacturers of pharmaceutical products need cer- Trichloroethane
hazard
tain information about the content of residual solvents
in excipients or drug substances in order to meet the 4.2 Solvents to Be Limited in Pharmaceutical
criteria of this guideline. The following statements are Products
given as acceptable examples of the information that Solvents in Table 2 should be limited in pharmaceu-
could be provided from a supplier of excipients or drug tical products because of their inherent toxicity. PDEs
substances to a pharmaceutical manufacturer. The sup- are given to the nearest 0.1 mg/day, and concentrations
plier might choose one of the following as appropriate: are given to the nearest 10 ppm. The stated values do
- Only Class 3 solvents are likely to be present. not reflect the necessary analytical precision of deter-
Loss on drying is less than 0.5%. mination. Precision should be determined as part of the
- Only Class 2 solvents X, Y, ... are likely to be validation of the method.
present. All are below the Option 1 limit. (Here
the supplier would name the Class 2 solvents Table 2. Class 2 solvents in pharmaceutical products.
represented by X, Y, ...)
PDE Concentration
- Only Class 2 solvents X, Y, ... and Class 3 sol- Solvent
(mg/day) limit (ppm)
vents are likely to be present. Residual Class 2
solvents are below the Option 1 limit and resid- Acetonitrile 4.1 410
ual Class 3 solvents are below 0.5%. Chlorobenzene 3.6 360
If Class 1 solvents are likely to be present, they Chloroform 0.6 60
should be identified and quantified. Cumene 0.7 70
"Likely to be present" refers to the solvent used in Cyclohexane 38.8 3880
the final manufacturing step and to solvents that are 1,2-Dichloroethene 18.7 1870
used in earlier manufacturing steps and not removed Dichloromethane 6.0 600
consistently by a validated process. 1,2-
1.0 100
If solvents of Class 2 or Class 3 are present at great- Dimethoxyethane
er than their Option 1 limits or 0.5%, respectively, they N,N-
10.9 1090
should be identified and quantified. Dimethylacetamide
N,N-
8.8 880
Dimethylformamide
4. LIMITS OF RESIDUAL SOLVENTS
1,4-Dioxane 3.8 380
2-Ethoxyethanol 1.6 160
4.1 Solvents to Be Avoided
Solvents in Class 1 should not be employed in the Ethyleneglycol 6.2 620
manufacture of drug substances, excipients, and drug Formamide 2.2 220
products because of their unacceptable toxicity or their Hexane 2.9 290
deleterious environmental effect. However, if their use Methanol 30.0 3000
is unavoidable in order to produce a drug product with 2-Methoxyethanol 0.5 50
a significant therapeutic advance, then their levels Methylbutyl ketone 0.5 50
should be restricted as shown in Table 1, unless other- Methylcyclohexane 11.8 1180
wise justified. 1,1,1-Trichloroethane is included in Ta- N-Methylpyrrolidone 48.4 4840
ble 1 because it is an environmental hazard. The stated Nitromethane 0.5 50
limit of 1500 ppm is based on a review of the safety Pyridine 2.0 200
data. Sulfolane 1.6 160
Tetrahydrofuran 7.2 720
Table 1. Class 1 solvents (solvents that should not be Tetralin 1.0 100
used in manufacture of pharmaceutical products). Toluene 8.9 890
1,1,2-
0.8 80
Trichloroethene
note 1)
Xylene 21.7 2170
note 1)
usually 60% m-xylene, 14% p-xylene, 9% o-
KP X 1853

xylene with 17% ethyl benzene Isopropyl ether Trifluoroacetic acid

4.3 Solvents with Low Toxic Potential Furthermore, for the Class 2 or Class 3 solvent is used
Solvents in Class 3 (shown in Table 3) may be re- prior to the last step of a manufacturing process of drug
garded as less toxic and of lower risk to human health. substances, it is not necessary to establish the specifi-
Class 3 includes no solvent known as a human health cation of the residual solvents of the drug substances,
hazard at levels normally accepted in pharmaceuticals. in case of complying with the requirements of the re-
However, there are no long-term toxicity or carcino- sidual solvents that are not remain in the final active
genicity studies for many of the solvents in Class 3. drug substances.
Available data indicate that they are less toxic in acute (Examples of the recommended data) the content of 3
or short-term studies and negative in genotoxicity stud- consecutive batches of residual solvents that is not
ies. It is considered that amounts of these residual sol- more than acceptable concentration limit and the data
vents of 50 mg per day or less (corresponding to 5000 of detection limit. When Class 2 solvents are used, they
ppm or 0.5% under Option 1) would be acceptable should be controlled either in a suitable intermediate or
without justification. Higher amounts may also be ac- in the management data of final active substances de-
ceptable provided they are realistic in relation to manu- pending on the batch scale.
facturing capability and good manufacturing practice
(GMP). 5. GLOSSARY
 Genotoxic Carcinogens: Carcinogens which pro-
Table 3. Class 3 solvents, which should be limited by duce cancer by affecting genes or chromosomes.
GMP or other quality-based requirements, in  LOEL: Abbreviation for lowest-observed effect
pharmaceutical products. level.
Acetic acid Heptane
 Lowest-Observed Effect Level: The lowest dose
Acetone Isobutyl acetate
of substance in a study or group of studies that
Anisole Isopropyl acetate
produces biologically significant increases in fre-
1-Butanol Methyl acetate
quency or severity of any effects in the exposed
2-Butanol 3-Methyl-1-butanol
humans or animals.
Butyl acetate Methylethyl ketone
 Modifying Factor: A factor determined by profes-
tert-Butylmethyl ether Methylisobutyl ketone
sional judgment of a toxicologist and applied to
Dimethyl sulfoxide 2-Methyl-1-propanol
bioassay data to relate that data safely to humans.
Ethanol Pentane
Ethyl acetate 1-Pentanol  Neurotoxicity: The ability of a substance to cause
adverse effects on the nervous system.
Ethyl ether 1-Propanol
Ethyl formate 2-Propanol  NOEL: Abbreviation for no-observed-effect level.
Formic acid Propyl acetate  No-Observed-Effect Level: The highest dose of
substance at which there are no biologically sig-
4.4 Solvents for which No Adequate Toxicological nificant increases in frequency or severity of any
Data was Found effects in the exposed humans or animals.
The following solvents (Table 4) may be of inter-  PDE: Abbreviation for permitted daily exposure.
est to manufacturers of excipients, drug substances, or  Permitted Daily Exposure: The maximum ac-
drug products. However, no adequate toxicological ceptable intake per day of residual solvent in
data on which to base a PDE was found. Manufacturers pharmaceutical products.
should supply justification for residual levels of these  Reversible Toxicity: The occurrence of harmful
solvents in pharmaceutical products. effects that are caused by a substance and which
disappear after exposure to the substance ends.
Table 4. Solvents for which no adequate toxicologi-  Strongly Suspected Human Carcinogen: A sub-
cal data was found. stance for which there is no epidemiological evi-
dence of carcinogenesis but there are positive
Methylisopropyl ke- genotoxicity data and clear evidence of carcino-
1,1-Diethoxypropane genesis in rodents.
tone
1,1-Dimethoxymethane Methyltetrahydrofuran  Teratogenicity: The occurrence of structural mal-
formations in a developing fetus when a sub-
2,2-Dimethoxypropane Petroleum ether
stance is administered during pregnancy.
Isooctane Trichloroacetic acid

APPENDIX 1. LIST OF SOLVENTS INCLUDED IN THE GUIDELINE

Solvent Other Names Classification


Acetic acid Ethanoic acid Class 3
Acetone 2-Propanone; Propan-2-one Class 3
1854 Monographs, Part II

Acetonitrile Class 2
Anisole Methoxybenzene Class 3
Benzene Benzol Class 1
1-Butanol n-Butyl alcohol Class 3
Butan-1-ol
2-Butanol sec-Butyl alcohol; Butan-2-ol Class 3
Butyl acetate Acetic acid butyl ester Class 3
tert-Butylmethyl ether 2-Methoxy-2-methyl- propane Class 3
Carbon tetrachloride Tetrachloromethane Class 1
Chlorobenzene Class 2
Chloroform Trichloromethane Class 2
Cumene Isopropylbenzene; (1-Methyl)ethylbenzene Class 2
Cyclohexane Hexamethylene Class 2
1,2-Dichloroethane sym-Dichloroethane; Ethylene dichloride; Ethylene chloride Class 1
1,1-Dichloroethene 1,1-Dichloroethylene; Vinylidene chloride Class 1
1,2-Dichloroethene 1,2-Dichloroethylene; Acetylene dichloride Class 2
Dichloromethane Methylene chloride Class 2
1,2-Dimethoxyethane Ethyleneglycol dimethyl ether; Monoglyme; Dimethyl Cellosolve Class 2
N,N- DMA Class 2
Dimethylacetamide
N,N- DMF Class 2
Dimethylformamide
Dimethyl sulfoxide Methylsulfinylmethane; Methyl sulfoxide; DMSO Class 3
1,4-Dioxane p-Dioxane; [1,4]Dioxane Class 2
Ethanol Ethyl alcohol Class 3
2-Ethoxyethanol Cellosolve Class 2
Ethyl acetate Acetic acid ethyl ester Class 3
Ethyleneglycol 1,2-Dihydroxyethane; 1,2-Ethanediol Class 2
Ethyl ether Diethyl ether; Ethoxyethane; 1,1’-Oxybisethane Class 3
Ethyl formate Formic acid ethyl ester Class 3
Formamide Methanamide Class 2
Formic acid Class 3
Heptane n-Heptane Class 3
Hexane n-Hexane Class 2
Isobutyl acetate Acetic acid isobutyl ester Class 3
Isopropyl acetate Acetic acid isopropyl ester Class 3
Methanol Methyl alcohol Class 2
2-Methoxyethanol Methyl Cellosolve Class 2
Methyl acetate Acetic acid methyl ester Class 3
3-Methyl-1-butanol Isoamyl alcohol; Isopentyl alcohol; 3-Methylbutan-1-ol Class 3
Methylbutyl ketone 2-Hexanone; Hexan-2-one Class 2
Methylcyclohexane Cyclohexylmethane Class 2
Methylethyl ketone 2-Butanone; MEK; Butan-2-one Class 3
Methylisobutyl ketone 4-Methylpentan-2-one; 4-Methyl-2-pentanone; MIBK Class 3
2-Methyl-1-propanol Isobutyl alcohol; 2-Methylpropan-1-ol Class 3
N-Methylpyrrolidone 1-Methylpyrrolidin-2-one; 1-Methyl-2-pyrrolidinone Class 2
Nitromethane Class 2
Pentane n-Pentane Class 3
1-Pentanol Amyl alcohol; Pentan-1-ol; Pentyl alcohol Class 3
1-Propanol Propan-1-ol; Propyl alcohol Class 3
2-Propanol Propan-2-ol; Isopropyl alcohol Class 3
Propyl acetate Acetic acid propyl ester Class 3
Pyridine Class 2
Sulfolane Tetrahydrothiophene 1,1-dioxide Class 2
Tetrahydrofuran Tetramethylene oxide; Oxacyclopentane Class 2
Tetralin 1,2,3,4-Tetrahydro-naphthalene Class 2
Toluene Methylbenzene Class 2
1,1,1-Trichloroethane Methylchloroform Class 1
KP X 1855

1,1,2-Trichloroethene Trichloroethene Class 2


Xylenenote) Dimethybenzene; Xylol Class 2
note)
usually 60% m-xylene, 14% p-xylene, 9% o-xylene with 17% ethyl benzene

APPENDIX 2. ADDITIONAL BACKGROUND EXPOSURE LIMITS


The Gaylor-Kodell method of risk assessment
2.1. Environmental Regulation of Organic Vola- (Gaylor, D. W. and Kodell, R. L.: Linear Interpola-
tile Solvents tion algorithm for low dose assessment of toxic sub-
Several of the residual solvents frequently stance. J Environ. Pathology, 4, 305, 1980) is ap-
used in the production of pharmaceuticals are listed propriate for Class 1 carcinogenic solvents. Only in
as toxic chemicals in Environmental Health Criteria cases where reliable carcinogenicity data are availa-
(EHC) monographs and the Integrated Risk Infor- ble should extrapolation by the use of mathematical
mation System (IRIS). The objectives of such models be applied to setting exposure limits. Expo-
groups as the International Programme on Chemical sure limits for Class 1 solvents could be determined
Safety (IPCS), the United States Environmental with the use of a large safety factor (i.e., 10,000 to
Protection Agency (USEPA), and the United States 100,000) with respect to the no-observed-effect lev-
Food and Drug Administration (USFDA) include el (NOEL). Detection and quantitation of these sol-
the determination of acceptable exposure levels. The vents should be by state-of-the-art analytical tech-
goal is protection of human health and maintenance niques.
of environmental integrity against the possible dele- Acceptable exposure levels in this guideline for
terious effects of chemicals resulting from long- Class 2 solvents were established by calculation of
term environmental exposure. The methods in- PDE values according to the procedures for setting
volved in the estimation of maximum safe exposure exposure limits in pharmaceuticals (Pharmacopeial
limits are usually based on long-term studies. When Forum, Nov-Dec 1989), and the method adopted by
long-term study data are unavailable, shorter term IPCS for Assessing Human Health Risk of Chemi-
study data can be used with modification of the ap- cals (Environmental Health Criteria 170, WHO,
proach such as use of larger safety factors. The ap- 1994). These methods are similar to those used by
proach described therein relates primarily to long- the USEPA (IRIS) and the USFDA (Red Book) and
term or life-time exposure of the general population others. The method is outlined here to give a better
in the ambient environment, i.e. ambient air, food, understanding of the origin of the PDE values. It is
drinking water and other media. not necessary to perform these calculations in order
to use the PDE values tabulated in Section 4 of this
2.2. Residual Solvents in Pharmaceuticals document.
Exposure limits in this guideline are estab- PDE is derived from the no-observed-effect level
lished by referring to methodologies and toxicity (NOEL), or the lowest-observed effect level (LOEL)
data described in EHC and IRIS monographs. How- in the most relevant animal study as follows:
ever, some specific assumptions about residual sol-
vents to be used in the synthesis and formulation of NOEL x Weight Adjustment (1)
PDE 
pharmaceutical products should be taken into ac- F1 x F2 x F3 x F4 x F5
count in establishing exposure limits. They are:
The PDE is derived preferably from a NOEL. If no
1) Patients (not the general population) use pharma- NOEL is obtained, the LOEL may be used. Modify-
ceuticals to treat their diseases or for prophylaxis to ing factors proposed here, for relating the data to
prevent infection or disease. humans, are the same kind of "uncertainty factors"
2) The assumption of life-time patient exposure is used in Environmental Health Criteria (Environ-
not necessary for most pharmaceutical products but mental Health Criteria 170, World Health Organiza-
may be appropriate as a working hypothesis to re- tion, Geneva, 1994), and "modifying factors" or
duce risk to human health. "safety factors" in Pharmacopeial Forum. The as-
3) Residual solvents are unavoidable components in sumption of 100% systemic exposure is used in all
pharmaceutical production and will often be a part calculations regardless of route of administration.
of drug products.
4) Residual solvents should not exceed recommend- The modifying factors are as follows:
ed levels except in exceptional circumstances. F1 = A factor to account for extrapolation between
5) Data from toxicological studies that are used to species
determine acceptable levels for residual solvents F1 = 5 for extrapolation from rats to humans
should have been generated using appropriate pro- F1 = 12 for extrapolation from mice to humans
tocols such as those described for example by F1 = 2 for extrapolation from dogs to humans
OECD, EPA, and the FDA Red Book. F1 = 2.5 for extrapolation from rabbits to hu-
mans
APPENDIX 3. METHODS FOR ESTABLISHING F1 = 3 for extrapolation from monkeys to hu-
1856 Monographs, Part II

mans adjustment for a lower body weight would be ap-


F1 = 10 for extrapolation from other animals to propriate.
humans As an example of the application of the equation (1),
F1 takes into account the comparative surface area: consider a toxicity study of acetonitrile in mice that
body weight ratios for the species concerned and for is summarized in Pharmeuropa, Vol. 9, No. 1, Sup-
man. Surface area (S) is calculated as: plement, April 1997, page S24. The NOEL is calcu-
lated to be 50.7 mg kg-1 day-1. The PDE for ace-
S = kM 0.67 (2) tonitrile in this study is calculated as follows:

in which M = body mass, and the constant k has 50.7 mg kg -1 day -1  50 kg


been taken to be 10. The body weights used in the PDE   4.2 mg day -1
12 10  5 11
equation are those shown below in the Table 1.
In this example,
F2 = A factor of 10 to account for variability F1 = 12 to account for the extrapolation from
between individuals mice to humans
A factor of 10 is generally given for all F2 = 10 to account for differences between indi-
organic solvents, and 10 is used consist- vidual humans
ently in this guideline. F3 = 5 because the duration of the study was
F3 = A variable factor to account for toxicity only 13 weeks
studies of short-term exposure F4 = 1 because no severe toxicity was encoun-
F3 = 1 for studies that last at least one half life- tered
time (1 year for rodents or rabbits; 7 years F5 = 1 because the no effect level was deter-
for cats, dogs and monkeys). mined
F3 = 1 for reproductive studies in which the
whole period of organogenesis is covered. Table 1. Values used in the calculations in this
F3 = 2 for a 6-month study in rodents, or a 3.5- guideline.
year study in non-rodents.
F3 = 5 for a 3-month study in rodents, or a 2- rat body weight 425 g
year study in non-rodents. pregnant rat body weight 330 g
F3 = 10 for studies of a shorter duration.
In all cases, the higher factor has been used for mouse body weight 28 g
study durations between the time points, e.g. a fac- pregnant mouse body weight 30 g
tor of 2 for a 9-month rodent study.
guinea pig body weight 500 g
F4 = A factor that may be applied in cases of severe
toxicity, e.g. non-genotoxic carcinogenicity, neuro- Rhesus monkey body weight 2.5 kg
toxicity or teratogenicity. In studies of reproductive rabbit body weight
toxicity, the following factors are used: 4 kg
(pregnant or not)
F4 = 1 for fetal toxicity associated with mater-
beagle dog body weight 11.5 kg
nal toxicity
F4 = 5 for fetal toxicity without maternal toxici- rat respiratory volume 290 L/day
ty
mouse respiratory volume 43 L/day
F4 = 5 for a teratogenic effect with maternal
toxicity rabbit respiratory volume 1440 L/day
F4 = 10 for a teratogenic effect without mater- guinea pig respiratory volume 430 L/day
nal toxicity
F5 = A variable factor that may be applied if the no- human respiratory volume 28,800 L/day
effect level was not established dog respiratory volume 9,000 L/day
When only an LOEL is available, a factor of up to
10 could be used depending on the severity of the monkey respiratory volume 1,150 L/day
toxicity. mouse water consumption 5 mL/day
The weight adjustment assumes an arbitrary adult
human body weight for either sex of 50 kg. This rat water consumption 30 mL/day
relatively low weight provides an additional safety rat food consumption 30 g/day
factor against the standard weights of 60 kg or 70 kg
that are often used in this type of calculation. It is
recognized that some adult patients weigh less than The equation for an ideal gas, PV = nRT, is used to
50 kg; these patients are considered to be accom- convert concentrations of gases used in inhalation
modated by the built-in safety factors used to de- studies from units of ppm to units of mg/L or mg/m3.
termine a PDE. If the solvent was present in a for- Consider as an example in which the rat reproduc-
mulation specifically intended for pediatric use, an tive toxicity study by inhalation of carbon tetrachlo-
ride (molecular weight 153.84) is summarized in
KP X 1857

Pharmeuropa, Vol. 9, No. 1, Supplement, April 1997, The degree of revalidation required depends on the
page S9. nature of the changes.
In addition, validation methods, other than those
1
n P 00 x 10 atm x 15 8 0 mg mol described in this guideline, may be used, although
the appropriateness of the alternative method has
RT 0 08 L atm 1 mol 1 x 8
15 mg to be justified.
1 8 mg L
5L
III. GLOSSARY
The relationship 1000 L = 1 m3 is used to convert to 1. Analytical Procedure
mg/ m3. The analytical procedure refers to the way of per-
forming the analysis. It should describe in detail
the steps necessary to perform each analytical test.
The analytical procedure includes but is not lim-
7. Guideline of Validation of ited to: the analyte, the sample, the reference
Analytical Procedures for standard, the standard reagents, the use of analyti-
cal instruments, the use of the calibration curve,
Pharmaceuticals the use of formulae for the calculation in the iden-
tification test, purity test or assay.
I. OBJECTIVE - The term ‘validation of analytical proce-
The purpose of this guideline is to provide the de- dure’ is defined as the process of confirming
tailed guidance on how to conduct the validation of that the analytical procedure for a quality
the analytical procedures necessary for application control test of pharmaceuticals is suitable for
(approval) for manufacture · import of pharmaceu- its intended use.
ticals · quasi-drugs according to notifications of - Identification Test refers to ensure the iden-
the Korean Food and Drug Administration, such as tity of an analyte, and generally compare the
‘Regulation of Pharmaceuticals Approval, Notifi- physicochemical characteristics (spectrum,
cation and Review’, and for quality control. information from chromatographic methods,
and chemical reactivity, etc) of a sample with
II. INTRODUCTION those of a reference material.
The objective of validation of an analytical pro- - Purity Test is to ensure that all the analytical
cedure for pharmaceuticals is to demonstrate that procedures performed allow an accurate
the procedure, applied in the quality control of the statement of the content of impurities of an
pharmaceutical, is suitable for the intended pur- analyte, i.e. related substances test, heavy
pose. metals, residual solvents content, etc. Testing
The purpose is to direct how to establish relevant for impurities can be either a quantification
validation parameters for each analytical procedure. test or a limit test for the impurity in a sample.
The scope of analytical procedures that are sub- - Assay (Content or Potency) is to provide an
jected to the present guideline is as follows: exact result which allows an accurate state-
1) Identification tests in the specification for ment on the content or potency of the analyte
pharmaceuticals in a sample. The assay represents a quantita-
2) Purity tests: quantitative tests and limit tests tive measurement of the major component(s)
for the impurity in a sample or other selected component(s) (i.e., stabilizer,
3) Quantification tests: assay for the active additive) in the drug substance or in the drug
component of drug substance or drug prod- product. The validation principles are also
uct, for other selected component(s) in drug applicable to assay for dissolution test.
products, for Uniformity of Dosage Units
test or for dissolution test 2. Specificity
The objective of the analytical procedure should Specificity is the ability to assess unequivocally
be clear since the procedure will govern the valida- the analyte in the presence of components, e.g.,
tion characteristics which need to be evaluated. impurities, degradation products, matrix, etc,
Typical validation parameters include specificity, which may be expected to be present. Lack of
accuracy, precision, detection limit, quantification specificity of an individual analytical procedure
limit, linearity, range, and robustness. Appropriate may be compensated by other supporting analyti-
validation parameters are selected and evaluated cal procedure(s).
depending on the objective of a given analytical
procedure. 3. Accuracy
Furthermore, revalidation may be necessary when The accuracy of an analytical procedure express-
there are change in the synthesis of the drug sub- es the closeness of agreement between the value
stance, change in the composition of the finished which is accepted either as a conventional true
product, and change in the analytical procedure. value or an accepted reference value and the value
1858 Monographs, Part II

found. ing normal usage.

4. Precision IV. VALIDATION OF ANALYTICAL PROCE-


The precision of an analytical procedure express- DURES
es the closeness of agreement (degree of scatter) For validation of analytical procedures, the vali-
between a series of measurements obtained from dation procedure and protocol most suitable for
homogeneous sample under the prescribed condi- their product must be first selected. Then, all data
tions. Precision may be considered at three levels: and validation parameters collected during the val-
repeatability, intermediate precision and reproduc- idation process are used to evaluate the appropri-
ibility. ateness. The relevant data collected during valida-
- Repeatability expresses the precision of tion and formulae used for calculating validation
measurements obtained under the same oper- parameters should be presented and justified as
ating conditions (same analyst, same labora- appropriate. The validation parameters applicable
tory, same equipment, same sample, etc) over to the identification test, the control of impurities
a short interval of time. Repeatability is also and the assay procedures is summarized in Appen-
termed intra-assay precision. dix 1.
- Intermediate Precision expresses within- Well-characterized reference materials, which
laboratories variations: different days, differ- documented purity as well as physicochemical and
ent analysts, different equipment, etc. biological characteristics, should be used through-
- Reproducibility expresses the precision of out the validation study. The degree of purity nec-
observed values (collaborative studies, usual- essary depends on the intended use.
ly applied to standardization of methodology)
obtained from a homogenous sample in dif- 1. Specificity
ferent laboratories. An investigation of specificity should be con-
ducted during the validation of identification tests,
5. Detection Limit the control of impurities and the assay procedures.
The detection limit of an individual analytical pro- The procedures used to demonstrate specificity
cedure is the lowest amount of analyte in a sample will depend on the intended objective of the ana-
which can be detected but not necessarily quanti- lytical procedure.
fied as an exact value. It is not always possible to demonstrate that an
analytical procedure is specific for a particular
6. Quantitation Limit analyte with complete discrimination. In this case,
The quantitation limit of an individual analytical a combination of two or more analytical proce-
procedure is the lowest amount of analyte in a dures is recommended to achieve the necessary
sample which can be quantitatively determined level of discrimination.
with suitable precision and accuracy. The quantita-
tion limit is a parameter of quantitative assays for A. Identification
low levels of compounds in sample matrices, and Suitable identification tests should be able to dis-
is used particularly for the determination of impu- criminate between compounds of closely related
rities and/or degradation products. structures which are likely to be present. The dis-
crimination of a procedure may be confirmed by
7. Linearity obtaining positive results, based on the comparison
The linearity of an analytical procedure is its abil- with a known reference material, from samples
ity to obtain test results which are directly propor- containing the analyte, coupled with negative re-
tional to the amount (concentration) of analyte in sults from samples which do not contain the
the sample. analyte. In addition, the identification test may be
applied to materials structurally similar to or close-
8. Range ly related to the analyte to confirm that a positive
The range of an analytical procedure is the inter- response is not obtained. The choice of such poten-
val between the upper and lower concentration tially interfering materials should be based on
(amounts) of analyte in the sample (including these sound scientific judgement with a consideration of
concentrations) for which it has been demonstrated the interferences that could occur.
that the analytical procedure has a suitable level of
precision, accuracy and linearity. B. Assay and Impurity Test(s)
For chromatographic procedures, representative
9. Robustness chromatograms should be presented to demon-
The robustness of an analytical procedure is a strate specificity and individual components
measure of its capacity to remain unaffected by should be appropriately labeled. Similar consid-
small, but deliberate variations in method parame- erations should be given to other separation tech-
ters. It provides an indication of its reliability dur- niques.
KP X 1859

Critical separations in chromatography should the range (see section 3) of the analytical proce-
be investigated at an appropriate level to demon- dure. It may be demonstrated directly on the drug
strate that the components are adequately sepa- substance (by dilution of a standard stock solution)
rated. For critical separations, specificity can be and/or separate weighings of synthetic mixtures of
demonstrated by the resolution of the two com- the drug product components, using the proposed
ponents which elute closest to each other. procedure. The latter aspect can be studied during
In cases where a non-specific assay is used, oth- investigation of the range.
er supporting analytical procedures should be Linearity should be evaluated by visual inspec-
used to demonstrate overall specificity. For ex- tion of a plot of signals as a function of analyte
ample, where a titration is adopted to assay the concentration or content. If there is a linear rela-
drug substance for release, the combination of the tionship, test results should be evaluated by appro-
assay and a suitable test for impurities can be priate statistical methods, for example, by calcula-
used. tion of a regression line by the method of least
The approach is similar for both assay and im- squares. In some cases, to obtain linearity between
purity tests. assays and sample concentrations, the test data
may need to be subjected to a mathematical trans-
(1) Impurities are available formation prior to the regression analysis. Data
For the assay, this should involve demonstration from the regression line itself may be helpful to
of the discrimination of the analyte in the pres- provide mathematical estimates of the degree of
ence of impurities and/or excipients; practically, linearity.
this can be done by spiking pure substances (drug The correlation coefficient, y-intercept, slope of
substance or drug product) with appropriate lev- the regression line and residual sum of squares
els of impurities and/or excipients and demon- should be submitted. A plot of the data should be
strating that the assay result is unaffected by the included. In addition, an analysis of the deviation
presence of these materials (by comparison with of the actual data points from the regression line
the assay result obtained on unspiked samples). may also be helpful for evaluating linearity.
For the impurity test, the discrimination may be Some analytical procedures, such as immunoas-
established by spiking drug substance or drug says, do not demonstrate linearity after any trans-
product with appropriate levels of impurities and formation. In this case, the analytical response
demonstrating the separation of these impurities should be described by an appropriate function of
individually and/or from other components in the the concentration (amountly or empiraclly) of the
sample matrix. concentration (or content) of an analyte in a sam-
ple.
(2) Impurities are not available For the establishment of linearity, a minimum of
If impurity or degradation products standards 5 concentrations is recommended. Other approach-
are unavailable, specificity may be demonstrated es should be justified.
by comparing the test results of samples contain-
ing impurities or degradation products to a se- 3. Range
cond well-characterized procedure. In this ap- The specified range is normally derived from lin-
proach, analytical procedure having known vali- earity studies and depends on the intended applica-
dation parameter includes pharmacopoeial meth- tion of the procedure. It is established by confirm-
od, other validated analytical procedure e.g.: ing that the analytical procedure provides an ac-
pharmacopoeial method of other validated analyt- ceptable degree of linearity, accuracy and precision
ical procedure (independent procedure). As ap- when applied to samples containing amounts of
propriate, this should include samples stored un- analyte within or at the extremes of the specified
der relevant stress conditions light, heat, humidity, range of the analytical procedure.
acid/base hydrolysis and oxidation. The following minimum specified ranges should
be considered:
- For the assay, the two results should be com-
pared. A. Assay of a drug substance or a drug product
- For the impurity tests, the impurity profiles Normally from 80 to 120 percent of the test con-
should be compared. centration.
B. Content uniformity
Peak purity tests may be useful to show that the Covering a minimum of 70 to 130 percent of the
analyte chromatographic peak is not attributable test concentration, unless a wider more appropriate
to more than one component (e.g., diode array, range, based on the nature of the dosage form such
mass spectrometry). as metered dose inhalers, is justified
C. Dissolution testing
2. Linearity ±20 % over the specified range indicated in disso-
A linear relationship should be evaluated across lution specification of the Specifications and Test
1860 Monographs, Part II

Procedures of the drug product. second well-characterized procedure,


For example, if the specifications for a controlled the accuracy of which is stated and/or
released product cover a region from 20%, after 1 defined (independent procedure).
hour, up to 90%, after 24 hours, the validated (c) Accuracy may be inferred once preci-
range would be 0-110% of the labeled content. sion, linearity and specificity have
D. Assay of related substance been established.
From the reporting level of an impurity to 120%
of the specification. B. Quantification of Impurities
For impurities known to be unusually potent or to Accuracy should be assessed on samples (drug
produce toxic or unexpected pharmacological ef- substance/drug product) spiked with known
fects, the detection/quantitation limit should be amounts of impurities.
commensurate with the level at which the impuri- In cases where it is impossible to obtain samples
ties must be controlled. For validation of impurity of certain impurities, and/or degradation products,
test procedures carried out during development, it it is considered acceptable to compare results ob-
may be necessary to consider the range around a tained by an independent procedure. The response
suggested (probable) limit. factor of the drug substance can be used.
E. If assay and purity are performed together as It should be clear how the individual or total im-
one test and only a 100% standard is used, lineari- purities are to be determined, e.g., weight/weight
ty should cover the range from the reporting level or area percent, in all cases with respect to the ma-
of the impurities to 120% of the assay specifica- jor analyte.
tion.
C. Recommended Data
4. Accuracy Accuracy should be assessed using a minimum of
Accuracy should be established across the speci- 9 determinations over a minimum of 3 concentra-
fied range of the analytical procedure. tion levels covering the specified range (e.g., 3
concentrations/3 replicates each of the total analyt-
A. Assay ical procedure).
(1) Drug Substance Accuracy should be reported as percent recovery
Several methods of determining accuracy are by the assay of known added amount of analyte in
available: the sample or as the difference between the mean
(a) When the purity is known and the accepted true value together with the con-
Application of an analytical procedure fidence intervals.
to an analyte of known purity (e.g.,
reference material); 5. Precision
(b) When other analytical procedure Validation of tests for assay and for quantitative
with the known accuracy is availa- determination of impurities includes an investiga-
ble tion of precision.
Comparison of the results of the pro-
posed analytical procedure with those A. Repeatability
of a second well characterized proce- Repeatability should be assessed using:
dure, the accuracy of which is stated (1) a minimum of 9 determinations covering the
and/or defined (independent proce- specified range for the procedure (e.g., 3 con-
dure); centrations/3 replicates each), or
(c) Accuracy may be inferred once preci- (2) a minimum of 6 determinations at 100% of
sion, linearity and specificity have the test concentration.
been established.
(2) Drug Product B. Intermediate precision
Several methods for determining accuracy are The extent to which intermediate precision should
available: be established depends on the circumstances under
(a) Application of the analytical proce- which the procedure is intended to be used. The
dure to synthetic mixtures of the drug applicant should establish the effects of random
product components to which known events on the precision of the analytical procedure.
quantities of the drug substance to be Typical variations to be studied include days, ana-
analyzed have been added; lysts, equipment, etc. The use of an experimental
(b) In cases where it is impossible to ob- design (matrix) is encouraged.
tain samples of all drug product com-
ponents, it may be acceptable either C. Reproducibility
1) to add known quantities of the analyte Reproducibility is assessed by means of an inter-
to the drug product, or laboratory trial. Reproducibility should be consid-
2) to compare the results obtained from a ered in case of the standardization of an analytical
KP X 1861

procedure, for instance, for inclusion of procedures A specific calibration curve should be stud-
in pharmacopoeias. These data are not part of the ied using samples containing an analyte in the
marketing authorization dossier. range of DL. The residual standard deviation
of a regression line or the standard deviation
D. Recommended Data of y-intercepts of regression lines may be
The standard deviation, relative standard deviation used as the standard deviation.
(coefficient of variation) and confidence interval
should be reported for each type of precision in- D. Recommended Data
vestigated. The detection limit and the method used for de-
termining the detection limit should be presented.
6. Detection limit If DL is determined based on visual evaluation or
Several approaches for determining the detection based on signal-to-noise ratio, the presentation of
limit are possible, depending on whether the pro- the relevant chromatograms is considered accepta-
cedure is a non-instrumental or instrumental. Ap- ble for justification.
proaches other than those listed below may be ac- In cases where an estimated value for the detec-
ceptable. tion limit is obtained by calculation or extrapola-
tion, this estimate may subsequently be validated
A. Based on Visual Evaluation by the independent analysis of a suitable number
Visual evaluation may be used for non- of samples known to be near or prepared at the de-
instrumental methods but may also be used with tection limit.
instrumental methods.
The detection limit is determined by the analysis 7. Quantification limit
of samples with known concentrations of analyte Several approaches for determining the quantifi-
and by establishing the minimum level at which cation limit are possible, depending on whether the
the analyte can be reliably detected. procedure is non-instrumental or instrumental.
Approaches other than those listed below may be
B. Based on Signal-to-Noise acceptable.
This approach can only be applied to analytical
procedures which exhibit baseline noise. Determi- A. Based on Visual Evaluation
nation of the signal-to-noise ratio is performed by Visual evaluation may be used for non-
comparing measured signals from samples with instrumental methods but may also be used with
known low concentrations of analyte with those of instrumental methods.
blank samples and establishing the minimum con- The quantification limit is generally determined
centration at which the analyte can be reliably de- by the analysis of samples with known concentra-
tected. A signal-to-noise ratio between 3 or 2:1 is tions of analyte and by establishing the minimum
generally considered acceptable for estimating the level at which the analyte can be quantified with
detection limit. acceptable accuracy and precision.

C. Based on the Standard Deviation of the Re- B. Based on Signal-to-Noise Approach


sponse and the Slope This approach can only be applied to analytical
The detection limit (DL) may be expressed as: procedures that exhibit baseline noise. Determina-
tion of the signal-to-noise ratio is performed by
DL = . * σ / S comparing measured signals from samples with
known low concentrations of analyte with those of
where blank samples and by establishing the minimum
σ = the standard deviation of the response concentration at which the analyte can be reliably
S = the slope of the calibration curve quantified. A typical signal-to-noise ratio is 10:1.

The slope S may be estimated from the calibra- C. Based on the Standard Deviation of the Re-
tion curve of the analyte. The estimate of σ sponse and the Slope
may be carried out in a variety of ways for ex- The quantification limit (QL) may be expressed
ample: as:
(1) Based on the Standard Deviation of the
Blank QL = 10 * σ / S
Measurement of the magnitude of analytical
background response is performed by analyz- where
ing an appropriate number of blank samples σ = the standard deviation of the response
and calculating the standard deviation of the- S = the slope of the calibration curve
se responses.
(2) Based on the Calibration Curve The slope S may be estimated from the calibra-
1862 Monographs, Part II

tion curve of the analyte. The estimate of σ - temperature,


may be carried out in a variety of ways for ex- - flow rate
ample:
(1) Based on Standard Deviation of the Blank 9. System suitability testing
Measurement of the magnitude of analytical System suitability testing is an integral part of
background response is performed by analyz- many analytical procedures. The tests are based on
ing an appropriate number of blank samples the concept that the equipment, electronics, analyt-
and calculating the standard deviation of the- ical operations and samples to be analyzed consti-
se responses. tute an integral system that can be evaluated as
(2) Based on the Calibration Curve such. System suitability test parameters to be es-
A specific calibration curve should be stud- tablished for a particular procedure depend on the
ied using samples, containing an analyte in type of procedure being validated. See General
the range of QL. The residual standard devia- Tests for additional information.
tion of a regression line or the standard devia-
tion of y-intercepts of regression lines may be Appendix 1) Validation characteristics applicable
used as the standard deviation. to analytical procedures
- signifies that this characteristic is not normal-
D. Recommended Data ly evaluated
The quantification limit and the method used for + signifies that this characteristic is normally
determining the quantification limit should be pre- evaluated
sented. (1) in cases where reproducibility (see glossary)
The limit should be subsequently validated by the has been performed, intermediate precision is
analysis of a suitable number of samples known to not needed
be near or prepared at the quantification limit. (2) lack of specificity of one analytical procedure
could be compressed by other supporting
8. Robustness analytical procedure(s)
The evaluation of robustness should be consid- (3) may be needed in some cases
ered during the development phase and depends on
the type of procedure under study. It should show
the reliability of an analysis with respect to delib-
erate variations in method parameters.
8. Isoelectric focusing
If measurements are susceptible to variations in
analytical conditions, the analytical conditions General Principles
should be suitably controlled or a precautionary Isoelectric focusing (IEF) is a method of electro-
statement should be included in the procedure. One phoresis that separates proteins according to their isoe-
consequence of the evaluation of robustness lectric points. Separation is carried out in a slab of
should be that a series of system suitability param- polyacrylamide or agarose gel that contains a mixture
eters (e.g., resolution test) is established to ensure of amphoteric electrolytes (ampholytes). When sub-
that the validity of the analytical procedure is jected to an electric field, the ampholytes migrate in
maintained whenever used. the gel to create a pH gradient. In some cases, gels
containing an immobilized pH gradient, prepared by
A. Typical variation factors common for all an- incorporating weak acids and bases to specific regions
alytical procedures of the gel network during the preparation of the gel,
- stability of analytical solutions, are used. When the applied proteins reach the gel frac-
- extraction time tion that has a pH that is the same as their isoelectric
point (pI), their charge is neutralized and migration
B. Typical variation factors for liquid chroma- ceases. Gradients can be made over various ranges of
tography pH, according to the mixture of ampholytes chosen.
- influence of variations of pH in a mobile
phase, Theoretical Aspects
- influence of variations in mobile phase com- When a protein is at the position of its isoelectric
position, point, it has no net charge and cannot be moved in a
- different columns (different lots or suppliers), gel matrix by the electric field. It may, however, move
- temperature, from that position by diffusion. The pH gradient forc-
- flow rate es a protein to remain in its isoelectric point position,
thus concentrating it; this concentrating effect is called
C. Typical variation factors for gas chromatog- “focusing”. The applied voltage is limited by the heat
raphy generated, which must be dissipated. The use of thin
- change in columns (different lots and/or sup- gels and an efficient cooling plate controlled by a
pliers), thermostatic circulator prevents the burning of the gel
whilst allowing sharp focusing. The separation is es-
KP X 1863

timated by determining the minimum pI difference The time required for completion of focusing in
(ΔpI), which is necessary to separate 2 neighboring thin-layer polyacrylamide gels is determined by plac-
bands: ing a colored protein (e.g. hemoglobin) at different
positions on the gel surface and by applying the elec-
D  (dpH / dx) tric field: the steady state is reached when all applica-
R  pI  3  tions give an identical band pattern. In some protocols
E  (d / dpH ) the completion of the focusing is indicated by the time
elapsed after the sample application. The IEF gel can
D: Diffusion coefficient of the protein be used as an identity test when the migration pattern
dpH/dx: pH gradient on the gel is compared to a suitable standard prepara-
E: Intensity of the electric field, in volts per centi- tion and IEF calibration proteins, the IEF gel can be
meter used as a limit test when the density of a band on IEF
-dµ/dpH: Variation of the solute mobility with the is compared subjectively with the density of bands
pH in the region close to the pI appearing in a standard preparation, or it can be used
as a quantitative test when the density is measured
ASSAY using a densitometer or similar instrumentation to
Type of
Testing for - dissolu- determine the relative concentration of protein in the
analytical
impurities tion bands subject to validation.
procedure
(meas-
Identifi-
uremen Apparatus
cation
quan t only) An apparatus for IEF consists of :
characteris- - content
tific limit A controllable generator for constant potential, current
tics / po-
ation and power. Potentials of 2500 V have been used and
tency are considered optimal under a given set of operating
Accuracy - + - + conditions. Supply of up to 30 W of constant power is
Precision recommended,
Repeata- - + - + A rigid plastic IEF chamber that contains a cooled
bility - +(1) - +(1) plate, of suitable material, to support the gel,
Interm.Pr + + + + A plastic cover with platinum electrodes that are con-
ecision - -(3) + - nected to the gel by means of paper wicks of suitable
Specifici- - + - - width , length and thickness , impregnated with solu-
ty(2) - + - + tions of anodic and cathodic electrolytes .
Detection - + - +
Limit Isoelectric Focusing in Polyacrylamide Gels: De-
Quantifica- tailed Procedure
tion Limit The following method is a detailed description of
Linearity an IEF procedure in thick polyacrylamide slab gels,
Range which is used unless otherwise stated in the mono-
graph.
Since D and -dµ/dpH for a given protein cannot
be altered, the separation can be improved by using a Preparation of the Gels
narrower pH range and by increasing R, the intensity Mould The mould (see Figure) is composed of
of the electric field. Resolution between protein bands a glass plate (A) on which a polyester film (B) is
on an IEF gel prepared with carrier ampholytes can be placed to facilitate handling of the gel, one or more
quite good. Improvements in resolution may be spacers (C), a second glass plate (D) and clamps to
achieved by using immobilized pH gradients where hold the structure together .
the buffering species, which are analogous to carrier 7.5% Polyacrylamide gel Dissolve 29.1 g of
ampholytes, are copolymerized within the gel matrix. acrylamide and 0.9 g of N,N’-methylenebisacrylamide
Proteins exhibiting pIs differing by as little as 0.02 pH in 100 mL of water. To 2.5 volumes of this solution,
units may be resolved using a gel prepared with carrier add the mixture of ampholytes specified in the mono-
ampholytes while immobilized pH gradients can re- graph and dilute to 10 volumes with water. Mix care-
solve proteins differing by approximately 0.001 pH fully and degas the solution.
units.

Practical Aspects
Special attention must be paid to sample charac-
teristics and/or preparation. Having salt in the sample
can be problematic and it is best to prepare the sample,
if possible, in de-ionized water or 2% ampholytes,
using dialysis or gel filtration if necessary.
1864 Monographs, Part II

procedure may be made subject to validation. These


include:
(1) the use of commercially available pre-cast gels and
of commercial staining and destaining kits,
D (2) the use of immobilized pH gradients,
C (3) the use of disk gels,
B (4) the use of gel cassettes of different dimensions,
A including ultra-thin (0.2 mm) gels,
(5) variations in the sample application procedure,
including different sample volumes or the use of
Preparation of the mould Place the polyester
sample application masks or wicks other than paper ,
film on the lower glass plate, apply the spacer, place
(6) the use of alternate running conditions, including
the second glass plate and fit the clamps. Place 7.5%
variations in the electric field depending on gel di-
polyacrylamide gel prepared before use on a magnetic
mensions and equipment, and the use of fixed mi-
stirrer, and add 0.25 volumes of a solution of ammo-
gration times rather than subjective interpretation of
nium persulfate (1 in 10) and 0.25 volumes of
band stability ,
N,N,N’,N’-tetramethylethylenediamine. Immediately
(7) the inclusion of a pre-focusing step,
fill the space between the glass plates of the mould
(8) the use of automated instrumentation,
with the solution.
(9) the use of agarose gels.
Method
Dismantle the mould and, making use of the pol-
Validation of Iso-Electric Focusing Procedures
yester film, transfer the gel onto the cooled support,
Where alternative methods to the detailed proce-
wetted with 2-3 mL of a suitable liquid, taking care to
dure are employed they must be validated. The follow-
avoid forming air bubbles. Prepare the test solutions
ing criteria may be used to validate the separation:
and reference solutions as specified in the monograph.
(1) formation of a stable pH gradient of desired char-
Place strips of paper for sample application, about 10
acteristics, assessed for example using colored pH
mm × 5 mm in size, on the gel and impregnate each
markers of known isoelectric points ,
with the prescribed amount of the test and reference
(2) comparison with the electropherogram provided
solutions. Also apply the prescribed quantity of a solu-
with the chemical reference substance for the prepa-
tion of proteins with known isoelectric points as pH
ration to be examined,
markers. In some protocols, the gel has precast slots
(3) any other validation criteria as prescribed in the
where a solution of the sample is applied instead of
monograph.
using impregnated paper strips. Cut 2 strips of paper
to the length of the gel and impregnate them with the
Specified Variations to the General Method
electrolyte solutions (acid for the anode and alkaline
Variations to the general method required for the
for the cathode). The compositions of the anode and
analysis of specific substances may be specified in
cathode solutions are given in the monograph. Apply
detail in monographs. These include:
these paper wicks to each side of the gel several mil-
(1) the addition of urea in the gel (3 mol/L concentra-
limeters from the edge. Fit the cover so that the elec-
tion is often satisfactory to keep protein in solution but
trodes are in contact with the wicks (respecting the
up to 8mol/L can be used): some proteins precipitate
anodic and cathodic poles). Proceed with the isoelec-
at their isoelectric point. In this case, urea is included
tric focusing by applying the electrical parameters
in the gel formulation to keep the protein in solution.
described in the monograph. Switch off the current
If urea is used, only fresh solutions should be used to
when the migration of the mixture of standard proteins
prevent carbamylation of the protein,
has stabilized. Using forceps, remove the sample ap-
(2) the use of alternative staining methods,
plication strips and the 2 electrode wicks. Immerse the
(3) the use of gel additives such as non-ionic deter-
gel in ‘fixing solution for isoelectric focusing in poly-
gents (e.g. octylglucoside) or zwitterionic detergents
acrylamide gel’. Incubate with gentle shaking at room
[e.g., 3-[(3- cholamido propyl)dimethylammonio]-1-
temperature for 30 minutes. Drain off the solution and
propane sulfonate (CHAPS) or 3-[(3-
add 00 mL of ‘destaining solution’. Incubate with
cholamidopropyl)dimethyl ammonio]-2-hydroxy-1-
shaking for 1hour. Drain the gel, add ‘coomassie
propanesulfate (CHAPSO)], and the addition of
staining TS’. Incubate for 30 minutes. Destain the gel
ampholyte to the sample, to prevent proteins from
by passive diffusion with ‘destaining solution’ until
aggregating or precipitating.
the bands are well visualized against a clear back-
Points to Consider
ground. Locate the position and intensity of the bands
Samples can be applied to any area on the gel,
in the electropherogram as prescribed in the mono-
but to protect the proteins from extreme pH environ-
graph.
ments samples should not be applied close to either
Variations to the Detailed Procedure (Subject to Vali-
electrode. During method development the analyst can
dation)
try applying the protein in 3 positions on the gel (i.e.
Where reference to the general method on isoe-
middle and both ends); the pattern of a protein applied
lectric focusing is made, variations in methodology or
KP X 1865

at opposite ends of the gel may not be identical. py. Optical microscopy is particularly useful for char-
A phenomenon known as ‘cathodic drift’, where acterizing particles that are not spherical. This method
the pH gradient decays over time, may occur if a gel is may also serve as a base for the calibration of faster
focused too long. Although not well understood, and more routine methods that may be developed.
electroendoosmosis and absorption of carbon dioxide Apparatus - Use a microscope that is stable and pro-
may be factors that lead to cathodic drift. Cathodic tected from vibration. The microscope magnification
drift is observed as focused protein migrating off the (product of the objective magnification, ocular magni-
cathode end of the gel. Immobilized pH gradients may fication, and additional magnifying components) must
be used to address this problem. be sufficient to allow adequate characterization of the
Efficient cooling (approximately 4 °C) of the bed smallest particles to be classified in the test specimen.
that the gel lies on during focusing is important. High The greatest numerical aperture of the objective
field strengths used during isoelectric focusing can should be sought for each magnification range. Polar-
lead to overheating and affect the quality of the fo- izing filters may be used in conjunction with suitable
cused gel. analyzers and retardation plates. Color filters of rela-
tively narrow spectral transmission should be used
Reagents and Solutions with achromatic objectives and are preferable with
Fixing solution for isoelectric focusing in poly- apochromats and are required for appropriate color
acrylamide gel Dissolve 35 g of 5-sulfosalicylic rendition in photomicrography. Condensers corrected
acid dihydrate and 100 g of trichloroacetic acid in for at least spherical aberration should be used in the
water to make 1000 mL. microscope substage and with the lamp. The numeri-
Coomassie staining TS Dissolve 125 mg of cal aperture of the substage condenser should match
coomassie brilliant blue R-250 in 100 mL of a mixture that of the objective under the condition of use, in the
of water, methanol and acetic acid (100) (5:4:1), and other words this is affected by the actual aperture of
filter. the condenser diaphragm and the presence of immer-
Destaining solution A mixture of water, metha- sion oils.
nol and acetic acid (100) (5:4:1). Adjustment - The precise alignment of all elements
of the optical system and proper focusing are essential.
The focusing of the elements should be done in ac-
cordance with the recommendations of the microscope
9. Particle Size manufacturer. Critical axial alignment is recommend-
Determination ed.
Illumination - A requirement for good illumination
Particle Size Determination is a method to deter- is a uniform and adjustable intensity of light over the
mine directly or indirectly morphological appearance, entire field of view; Kohler illumination is preferred.
shape, size and its distribution of powdered pharma- With colored particles, choose the color of the filters
ceutical drugs and excipients to examine their used so as to control the contrast and detail of the im-
micromeritic properties. Optical microscopy or analyt- age.
ical sieving method is used according to the measuring
purpose and the properties of the test specimen. Visual Characterization - The magnification and
numerical aperture should be sufficiently high to allow
Method 1. Optical Microscopy adequate resolution of the images of the particles to be
The optical microscopy is used to observe the mor- characterized. Determine the actual magnification
phological appearance and shape of individual particle using a calibrated stage micrometer to calibrate an
either directly with the naked eye or by using a micro- ocular micrometer. Errors can be minimized if the
scopic photograph, in order to measure the particle magnification is sufficient that the image of the parti-
size. The particle size distribution can also be deter- cle is at least 10 ocular divisions. Each objective must
mined by this method. It is also possible with this be calibrated separately. The ocular scale, the stage
method to measure the size of the individual particle micrometer scale and the ocular scale should be
even then different kinds of particles mingle if they aligned to calibrate. In this way, a precise determina-
are optically distinguishable. Data processing tech- tion of the distance between ocular stage divisions can
niques, such as image analysis, can be useful for de- be made.
termining the particle size distribution. When the particle size is measured, an ocular mi-
This method for particle characterization can gener- crometer is inserted at the position of the ocular dia-
ally be applied to particles not less than 1 μm. The phragm, and a calibrated stage micrometer is placed at
lower limit is imposed by the resolving power of the the center of the microscope stage and fixed in place.
microscope. The upper limit is less definite and is de- The ocular is attached to the lens barrel and adjusted
termined by the increased difficulty associated with to the focus point of the stage micrometer scale. Then,
the characterization of large particles. Various alterna- the distance between the scales of the two microme-
tive techniques are available for particle characteriza- ters is determined, and the sample size equivalent 1
tion, outside the applicable range of optical microsco- division of the ocular scale is calculated using the fol-
1866 Monographs, Part II

lowing formula: the powder to be examined. Count all the particles


having a maximum dimension greater than the pre-
The particle size equivalent 1 division on the ocular scribed size limit. The size limit and the permitted
scale (m) = Length on the stage micrometer number of particles exceeding the limit are defined for
(m)/Number of scale divisions on the ocular mi- each substance.
crometer Particle Size Characterization - The measurement
of particle size varies in complexity depending on the
The stage micrometer is removed and the test speci- shape of the particle and the number of particles char-
men is placed on the microscope stage. After adjusting acterized must be sufficient to insure an acceptable
the focus, the particle sizes are determined from the level of uncertainty in the measured parameters1). For
number of scale divisions read through the ocular. irregular particles, a variety of definitions of particle
Several different magnifications may be necessary to size exist. In general, for irregularly shaped particles,
characterize materials having a wide particle size dis- characterization of particle size must also include in-
tribution. formation on the type of diameter measured as well as
Photographic Characterization - If particle size is information on particle shape. Several commonly used
to be determined by photographic methods, take care measurements of particle size are defined below (see
to ensure that the object is sharply focused at the plane Fig. 1):
of the photographic emulsion. Determine the actual
magnification by photographing a calibrated stage
micrometer, using photographic film of sufficient
speed, resolving power, and contrast. Exposure and
processing should be identical for photographs of both
the test specimen and the determination of magnifica-
tion. The apparent size of a photographic image is
influenced by the exposure, development, and printing
processes as well as by the resolving power of the
microscope.
Preparation of the Mount - The mounting medi-
um will be selected according to the physical proper-
ties of the test specimen. Sufficient, but not excessive,
contrast between the specimen and the mounting me-
dium is required to ensure adequate detail of the spec- Fig. 1 Commonly used measurements of particle size
imen edge. The particles should rest in one plane and
be adequately dispersed to distinguish individual par- Feret's Diameter - The distance between imaginary
ticles of interest. Furthermore, the particles must be parallel lines tangent to a randomly oriented particle
representative of the distribution of sizes in the mate- and perpendicular to the ocular scale.
rial and must not be altered during preparation of the Martin's Diameter - The diameter of the particle at
mount. Selection of the mounting medium must in- the point that divides a randomly oriented particle into
clude a consideration of the analyte solubility. two equal projected areas.
Crystallinity characterization - The crystallinity Projected area Diameter - The diameter of a circle
of a material may be characterized to determine com- that has the same projected are as the particle.
pliance with the crystallinity requirement where stated Length - The longest dimension from edge to edge of
in the individual monograph of a drug substance. Un- a particle oriented parallel to the ocular scale.
less otherwise specified in the individual monograph, Width - The longest dimension of the particle meas-
mount a few particles of the specimen in mineral oil ured at right angles to the length.
on a clean glass slide. Examine the mixture using a
polarizing microscope: the particles show birefrin- Particle Shape Characterization - For irregularly
gence (interference colors) and extinction positions shaped particles, characterization of particle size must
when the microscope stage is revolved. also include information on particle shape. The homo-
Limit Test of Particle Size by Microscopy - geneity of the powder should be checked using appro-
Weigh a suitable quantity of the power to be examined priate magnification. The following defines some
(for example, 10 to 100 mg), and suspend it in 10 mL commonly used descriptors of particle shape (see Fig.
of a suitable medium in which the power does not 2):
dissolve, adding, if necessary, a wetting agent. A ho-
mogeneous suspension of particles can be maintained
by suspending the particles in a medium of similar or
matching density and by providing adequate agitation.
Introduce a portion of the homogeneous suspension
into a suitable counting cell, and scan under a micro-
scope an area corresponding to not less than 10 g of
KP X 1867

When using a woven sieve cloth, the sieving will es-


sentially sort the particles by their intermediate size
dimension (i.e., breadth or width). Mechanical sieving
is most suitable where the majority of the particles are
larger than about 75 m. For smaller particles, the
light weight provides insufficient force during sieving
to overcome the surface forces of cohesion and adhe-
sion that cause the particles to stick to each other and
to the sieve, and thus cause particles that would be
expected to pass through the sieve to be retained. For
such materials other means of agitation such as air-jet
Fig. 2 Commonly used descriptions of particle shape sieving or sonic sifting may be more appropriate.
Nevertheless, sieving can sometimes be used for some
Acicular - Slender, needle-like particle of similar powders or granules having median particle sizes
width and thickness. smaller than 75 m where the method can be validated.
Columnar - Long, thin particle with a width and In pharmaceutical terms, sieving is usually the method
thickness that are greater than those of an acicular of choice for classification of the coarser grades of
particle. single powders or granules. It is a particularly attrac-
Flake - Thin, flat particle of similar length and width. tive method in that powders and granules are classi-
Plate - Flat particles of similar length and width but fied only on the basis of particle size, and in most cas-
with greater thickness than flakes. es the analysis can be carried out in the dry state.
Lath - Long, thin, and blade-like particle Among the limitations of sieving method are the
Equant - Particles of similar length, width and thick- need for an appreciable amount of sample (normally at
ness; both cubical and spherical particles are included. least 25 g, depending on the density of the powder or
granule, and the diameter of test sieves) and difficulty
General Observations - A particle is generally in sieving oily or other cohesive powders or granules
considered to be the smallest discrete unit. A particle that tend to clog the sieve openings. The method is
may be a liquid or semisolid droplet; a single crystal essentially a two-dimensional estimate of size because
or polycrystalline; amorphous or an agglomerate. Par- passage through the sieve aperture is frequently more
ticles may be associated. This degree of association dependent on maximum width and thickness than on
may be described by the following terms: length.
Lamellar - Stacked plates. This method is intended for estimation of the total
Aggregate - Mass of adhered particles. particle size distribution of a single material. It is not
Agglomerate - Fused or cemented particles. intended for determination of the proportion of parti-
Conglomerate - Mixture of two or more types of par- cles passing or retained on one or two sieves.
ticles. Estimate the particle size distribution as described
Spherulite - Radical cluster. under Dry Sieving Method, unless otherwise specified
Drusy - Particle covered with tiny particles. in the individual monograph. Where difficulty is expe-
Particle condition may be described by the following rienced in reaching the endpoint (i.e., material does
terms not readily pass through the sieves) or when it is nec-
Edges - Angular, rounded, smooth, sharp, fractured. essary to use the finer end of the sieving range (below
Optical - Color (using proper color balancing filters), 75 m), serious consideration should be given to the
transparent, translucent, opaque. use of an alternative particle-sizing method.
Defects - Occlusions, inclusions. Sieving should be carried out under conditions that
Surface characteristics may be described as: do not cause the test sample to gain or lose moisture.
Cracked - Partial split, break, or fissure. The relative humidity of the environment in which the
Smooth - Free of irregularities, roughness, or projec- sieving is carried out should be controlled to prevent
tions. moisture uptake or loss by the sample. In the absence
Porous - Having openings or passageways. of evidence to the contrary, analytical test sieving is
Rough - Bumpy, uneven, not smooth. normally carried at ambient humidity. Any special
Pitted - Small indentations. conditions that apply to a particular material should be
detailed in the individual monograph.
Method 2. Analytical Sieving Method Principles of Analytical Sieving - Analytical test
The analytical sieving method is a method to esti- sieves are constructed from a woven-wire mesh,
mate the particle size distribution of powered pharma- which is of simple weave that is assumed to give near-
ceutical drugs by sieving. The particle size determined ly square apertures and is sealed into the base of an
by this method is shown as the size of a minimum open cylindrical container. The basic analytical meth-
sieve opening through which the particle passes. od involves stacking the sieves on top of one another
Sieving is one of the oldest methods of classifying in ascending degrees of coarseness, and then placing
powders and granules by particle size distribution. the test powder on the top sieve.
1868 Monographs, Part II

The nest of sieves is subjected to a standardized peri- imen of 10 to 25 g is available, smaller diameter test
od of agitation, and then the weight of material re- sieves conforming to the same mesh specifications
tained on each sieve is accurately determined. The test (table 1) may be substituted, but the endpoint must be
gives the weight percentage of powder in each sieve re-determined. The use of test samples having a small-
size range. er mass (e. g. down to 5 g) may be needed. For mate-
This sieving process for estimating the particle size rials with low apparent particle density, or for materi-
distribution of a single pharmaceutical powder is gen- als mainly comprising particles with a highly iso-
erally intended for use where at least 80 % of the par- diametrical shape, specimen weights below 5 g for a
ticles are larger than 75 m. The size parameter in- 200 mm screen may be necessary to avoid excessive
volved in determining particle size distribution by blocking of the sieve. During validation of a particular
analytical sieving is the length of the side of the min- sieve analysis method, it is expected that the problem
imum square aperture through which the particle will of sieve blocking will have been addressed.
pass. If the test material is prone to picking up or losing
significant amounts of water with varying humidity,
TEST SIEVES the test must be carried out in an appropriately con-
Unless otherwise specified in the monograph, use the trolled environment. Similarly, if the test material is
sieves listed in the Table 1. known to develop an electrostatic charge, careful ob-
Sieves are selected to cover the entire range of parti- servation must be made to ensure that such charging is
cle sizes present in the test specimen. A nest of sieves not influencing the analysis. An antistatic agent, such
having a 2 progression of the area of the sieve as colloidal silicon dioxide and/or aluminum oxide,
openings is recommended. The nest of sieves is as- may be added at a 0.5 percent (m/m) level to minimize
sembled with the coarsest screen at the top and the this effect. If both of the above effects cannot elimi-
finest at the bottom. Use micrometers or millimeters in nated, an alternative particle-sizing technique must be
denoting test sieve openings. [Note - Mesh numbers selected.
are provided in the table for conversion purposes only.] Agitation Methods - Several different sieve and
Test sieves are made from stainless steel or, less pref- powder agitation devices are commercially available,
erably, from brass or other suitable non-reactive wire. all of which may be used to perform sieve analyses.
However, the different methods of agitation may give
Calibration and recalibration of test sieves is in ac- different results for sieve analyses and endpoint de-
cordance with the most current edition of ISO 3310- terminations because of the different types and magni-
12). Sieves should be carefully examined for gross tude of the forces acting on the individual particles
distortions and fractures, especially at their screen under test. Methods using mechanical agitation or
frame joints, before use. Sieves may be calibrated op- electromagnetic agitation, and that can include either a
tically to estimate the average opening size, and open- vertical oscillation or a horizontal circular motion, or
ing variability, of the sieve mesh. Alternatively, for tapping or a combination of both tapping and horizon-
the valuation of the effective opening of test sieves in tal circular motion are available. Entrainment of the
the size range of 212 to 850 m, Standard Glass particles in an air stream may also be used. The results
Spheres are available. Unless otherwise specified in must indicate which agitation method was used and
the individual monograph, perform the sieve analysis the agitation parameters used (if they can be varied).
at controlled room temperature and at ambient relative End point Determination - The test sieving analy-
humidity. sis is complete when the weight on any of the test
Cleaning Test Sieves - Ideally, test sieves should sieves does not change by more than 5 % or 0.1 g (10 %
be cleaned using only an air jet or a liquid stream. If in the case of 76 mm sieves) of the previous weight on
some apertures remain blocked by test particles, care- that sieve. If less than 5 % of the total specimen
ful gentle brushing maybe used as a last resort. weight is present on a given sieve, the endpoint for
Test Specimen - If the test specimen weight is not that sieve is increased to a weight change of not more
given in the monograph for a particular material, use a than 20 % of the previous weight on that sieve. If
test specimen having a weight between 25 and 100 g, more than 50 % of the total specimen weight is found
depending on the bulk density of the material, and test on any one sieve, unless this is indicated in the mono-
sieves having a 200 mm diameter. For 76 mm sieves graph, the test should be repeated, but with the addi-
the amount of material that can be accommodated is tion to the sieve nest of a more coarse sieve intermedi-
approximately 1/7th that which can be accommodated ate between that carrying the excessive weight and the
on a 200 mm sieve. Determine the most appropriate next coarsest sieve in the original nest, i.e., addition of
weight for a given material by test sieving accurately the ISO series sieve omitted from the nest of sieves.
weighed specimens of different weights, such as 25,
50, and 100 g, for the same time period on a mechani- SIEVING METHODS
cal shaker. [Note - If the test results are similar for the
25 g and 50 g specimens, but the 100 g specimen Mechanical agitation
shows a lower percentage through the finest sieve, the Dry Sieving Method - Tare each test sieve to the
100-g specimen size is too large.] Where only a spec- nearest 0.1 g. Place an accurately weighed quantity of
KP X 1869

test specimen on the top (coarsest) sieve, and replace In the sonic sifting method a nest of sieves is used,
the lid. Agitate the nest of sieves for 5 minutes. Then and the test specimen is carried in a vertically oscillat-
carefully remove each from the nest without loss of ing column of air that lifts the specimen and then car-
material. If there is some fine powder on the down ries it back against the mesh openings at a given num-
surface of each sieve, take if off by the brush gently, ber of pulses per minute. It may be necessary to lower
and combine it with the sieve fraction retained on each the sample amount to 5 g, when sonic shifting is em-
next down sieve. Reweigh each sieve, and determine ployed.
the weight of material on each sieve. Determine the The air jet sieving and sonic sieving methods may be
weight of material in the collecting pan in a similar useful for powders or granules when mechanical siev-
manner. Reassemble the nest of sieves, and agitate for ing techniques are incapable of giving a meaningful
5 minutes. Remove and weigh each sieve as previous- analysis.
ly described. Repeat these steps until the endpoint These methods are highly dependent upon proper
criteria are met (see Endpoint Determination under dispersion of the powder in the air current. This re-
Test Sieves). Upon completion of the analysis, recon- quirement may be hard to achieve if the method is
cile the weights of material. Total losses must not ex- used at the lower end of the sieving range (i. e., below
ceed 5 % of the weight of the original test specimen. 75m), when the particles tend to be more cohesive,
Repeat the analysis with a fresh specimen, but using and especially if there is any tendency for the material
a single sieving time equal to that of the combined to develop an electrostatic charge. For the above rea-
times used above. Confirm that this sieving time con- sons endpoint determination is particularly critical,
forms to the requirements for endpoint determination. and it is very important to confirm that the oversize
When this endpoint has been validated for a specific material comprises single particles and is not com-
material, then a single fixed time of sieving may be posed of aggregates.
used for future analyses, providing the particle size
distribution falls within normal variation. INTERPRETATION
If there is evidence that the particles retained on any The raw data must include the weight of test speci-
sieve are aggregates rather than single particles, the men, the total sieving time, and the precise sieving
use of mechanical dry sieving is unlikely to give good methodology and the set values for any variable pa-
reproducibility, a different particle size analysis meth- rameters, in addition to the weights retained on the
od should be used. individual sieves and in the pan. It may be convenient
to convert the raw data into a cumulative weight dis-
Air Entrainment Methods tribution in terms of a cumulative weight undersize,
Air jet and Sonic Shifter Sieving - Different types the range of sieves used should include a sieve
of commercial equipment that use a moving air cur- through which all the material passes. If there is evi-
rent are available for sieving. A system that uses a dence on any of the test sieves that the material re-
single sieve at a time is referred to as air jet sieving. It maining on it is composed of aggregates formed dur-
uses the same general sieving methodology as that ing the sieving process, the analysis is invalid.
described under the Dry Sieving Method, but with a
1)
standardized air jet replacing the normal agitation Additional information on particle size measurement,
mechanism. It requires sequential analyses on individ- sample size, and data analysis is available, for exam-
ual sieves starting with the finest sieve to obtain a par- ple, in ISO 9276.
2)
ticle size distribution. Air jet sieving often includes the International Organization for Standardization (ISO)
use of finer test sieves than used in ordinary dry siev- Specification ISO 3310-1; Test sieves-Technical re-
ing. This technique is more suitable where only over- quirements and testing
size or undersize fractions are needed.

ISO Nominal Aperture Recommended


US Sieve European Korean
USP
Principal sizes Supplementary sizes No. Sieve No. Sieve No.
Sieves(mesh)
R 20/3 R 20 R 40/3
11.20 mm 11.20 mm 11.20 mm 11200
10.00 mm
9.50 mm
9.00 mm
8.00 mm 8.00 mm 8.00 mm
7.10 mm
6.70 mm
6.30 mm
5.60 mm 5.60 mm 5.60 mm 5600 3.5
5.00 mm
4.75 mm 4
1870 Monographs, Part II

4.50 mm
4.00 mm 4.00 mm 4.00 mm 5 4000 4000 4.7
3.55 mm
3.35 mm 6 5.5
3.15 mm
2.80 mm 2.80 mm 2.80 mm 7 2800 2800 6.5
2.50 mm
2.36 mm 8 7.5
2.24 mm
2.00 mm 2.00 mm 2.00 mm 10 2000 2000 8.6
1.80 mm
1.70 mm 12 10
1.60 mm
1.40 mm 1.40 mm 1.40 mm 14 1400 1400 12
1.25 mm
1.18 mm 16 14
1.12 mm
1.00 mm 1.00 mm 1.00 mm 18 1000 1000 16
900 m
850 m 20 18
800 m
710 m 710 m 710 m 25 710 710 22
630 m
600 m 30 26
560 m
500 m 500 m 500 m 35 500 500 30
450 m
425 m 40 36
400 m
355 m 355 m 355 m 45 355 355 42
315 m
300 m 50 50
280 m
250 m 250 m 250 m 60 250 250 60
224 m
212 m 70 70
200 m
180 m 180 m 180 m 80 180 180 83
160 m
150 m 100 100
140 m
125 m 125 m 125 m 120 125 125 119
112 m
106 m 140 140
100 m
90 m 90 m 90 m 170 90 90 166
80 m
75 m 200 200
71 m
63 m 63 m 63 m 230 63 63 235
56 m
53 m 270 282
50 m
45 m 45 m 45 m 325 45 45 330
40 m
38 m 38 391
KP X 1871

10. Powder Particle Density out between 15 and 30 C, and the temperature must
not vary by more than 2 C during the course of
Determination measurement.
Firstly, weigh the mass of the test cell and record it.
The Powder Particle Density Determination is a After weighing out the amount of the sample as de-
method to determine particle density of powdered scribed in the individual monograph and placing it in
pharmaceutical drugs or raw materials of drugs, and the test cell, seal the cell in the pycnometer. Secondly,
the gas displacement pycnometer is generally used. introduce the measurement gas (helium) into the test
The powder density by this method is determined with cell, in order to remove volatile contaminants in the
an assumption that the volume of the gas displaced by powder. If necessary, keep the sample powder under
the powder in a closed system is equal to the volume reduced pressure to remove the volatile contaminants
of the powder. The bulk density at loose packing or in advance and use it as the test sample for measure-
the tapped density at tapping express the apparent ment.
densities of the powder, since interparticular void vol- Open the valve which connects the reference cell
ume of the powder is considered to be a part of the with the test cell, confirm with manometer that the
volume of the powder. On the contrary, the pressure inside the system is stable, and then read the
pycnometric particle density expresses the powder system reference pressure (Pr). Secondly, close the
density nearly equal to the crystal density, since the valve that connects to the two cells, and introduce the
volume of the powder, that is deducted with void vol- measurement gas into the test cell to achieve positive
ume of open pores accessible to gas, is counted. pressure. Confirm with the manometer that the pres-
Powder particle density is expressed in mass per sure inside is stable, and then read the initial pressure
unit volume (kg/m3), and generally expressed in g/cm3. (Pi). Open the valve to connect test cell with the refer-
ence cell. After confirming that the indicator of the
Apparatus manometer is stable, read the final pressure (Pf). Cal-
The schematic diagram of particle density appa- culate the sample volume (Vs) with the following
ratus for gas displacement pycnometric measurement equation.
is shown in Figure. The apparatus consists of a test
cell in which the sample is placed, a reference cell and Vr
Vs  Vc 
a manometer. Pi  Pr
1
Generally, helium is used as the measurement gas. Pf  Pr
The apparatus has to be equipped with a system capa-
ble of pressuring the test cell to the defined pressure
Vr: Reference cell volume (cm3)
through the manometer.
Vc: Test cell volume (cm3)
Calibration of apparatus The volumes of the
Vs: Sample volume (cm3)
test cell (Vc) and the reference cell (Vr) must be accu-
Pi: Initial pressure (kPa)
rately determined to the nearest 0.001 cm3. In order to
Pf: Final pressure (kPa)
assure accuracy of the results of volume obtained,
Pr: Reference pressure (kPa)
calibration of the apparatus is carried out as follows
using a calibration ball of known volume for particle
Repeat the measurement sequence for the same
density measurement. The final pressure (Pf) are de-
powder sample until consecutive measurements of the
termined for the initial empty test cell followed by the
sample volume agree to within 0.5 %, and calculate
test cell placed with the calibration ball for particle
the mean of sample volumes (Vs). Finally, unload the
density measurement in accordance with the proce-
test cell, weigh the mass of test cell, and calculate the
dures, and Vc and Vr are calculated using the equation
final sample mass by deducing the empty cell mass
described in the section of Procedure. Calculation can
from the test cell mass. The powder particle density ρ
be made taking into account that the sample volume
is calculated by the following equation.
(Vs) is zero in the first run.
m
Vr: Reference cell volume (cm3) 
Vc: Test cell volume (cm3) Vs
Vs: Sample volume (cm3)
M : Manometer (cm3) ρ: Powder particle density (g/ cm3)
m: Final sample mass (g)
Figure. Schematic diagram of a gas Vs: Sample volume (cm3)
displacement pycnometer used to
determine powder particle density

Procedure
The measurement of the particle density is carried
1872 Monographs, Part II

These test microorganisms are representative of


11. Preservatives-Effectiveness those that might be found in the environment in which
Tests the product is manufactured, used or stored, and they
are also recognized as opportunistic pathogens. In
The purpose of the Preservatives-Effectiveness addition to these strains designated as test microorgan-
Tests is to assess microbiologically the preservative isms, it is further recommended to use strains that
efficacy, either due to the action of product compo- might contaminate the product and grow on or in it,
nents themselves or any added preservatives for mul- depending on its characteristics. For the test microor-
tiple containers. The efficacy of the preservatives is ganisms received from coordinated collections of mi-
assessed by direct inoculation and mixing of the test croorganisms, one passage is defined as the transfer of
strains in the product, and titration of survival of the microorganisms from an established culture to a fresh
test strains with time. medium, and microorganisms subjected to not more
Preservatives must not be used solely to comply than 5 passages should be used for the tests. Single-
with GMP for drugs or to reduce viable aerobic counts. strain challenges rather than mixed cultures should be
In fact, preservatives themselves are toxic substances. used. The test strains can be harvested by growth on
Therefore, preservatives must not be added to prod- solid agar or liquid media.
ucts in amounts which might jeopardize the safety of Culture on agar plate media: Inoculate each of
human beings, and consideration must be given to the five test strains on the surface of agar plates or
minimizing the amounts of preservatives used. These agar slants. For growth of bacteria, use Soybean-
tests are commonly used to verify that products main- Casein Digest Agar Medium, and for yeasts and
tain their preservative effectiveness at the design moulds, use Sabouraud Agar, Glucose-Peptone Agar
phase of formulation or in the case of periodic moni- or Potato Dextrose Agar Medium. Incubate bacterial
toring. Although these tests are not performed for lot cultures at 30 C to 35 C for 18 to 24 hours, the cul-
releasing testing, the efficacy of the preservative pre- ture of C. albicans at 20 C to 25 C for 40 to 48
sent in the product packaged in the final containers hours and the culture of A. niger at 20 C to 25 C for
should be verified throughout the entire dating period. one week or until good sporulation is obtained. Har-
vest these cultured cells aseptically using a platinum
1. Products and Their Categories loop, etc. Suspend the collected cells in sterile physio-
The products have been divided into two catego- logical saline or in 0.1 % peptone water and adjust the
ries for these tests. Category I products are those made viable cell count to about 108 microorganisms per mL.
with aqueous bases or vehicles, and Category II prod- In the case of A. niger, suspend the cultured cells in
ucts are those made with nonaqueous bases or vehicles. sterile physiological saline or 0.1 % peptone water
Oil-in-water emulsions are considered Category I containing 0.05 w/v% of polysorbate 80 and adjust the
products, and water-in-oil emulsions Category II spore count to about 108 per mL. Use these suspen-
products. Category I is further divided into three sub- sions as the inocula.
types depending on the dosage form. Liquid cultures: After culturing each of the four
Category IA: Injections and other sterile strains except for A. niger in a suitable medium, re-
parenterals move the medium by centrifugation. Wash the cells in
Category IB: Nonsterile parenterals sterile physiological saline or 0.1 % peptone water and
Category IC: Oral products made with aqueous resuspend them in the same solution with the viable
bases (including syrup products to be dissolved or cell or spore count of the inoculum adjusted to about
suspended before use) 108 per mL.
Category II: All the dosage forms listed under Cat- When strains other than the five listed above are
egory I made with nonaqueous bases or vehicles. cultured, select a culture medium suitable for growth
of the strain concerned. The cell suspension may also
2. Test Microorganisms and Culture Media be prepared by a method suitable for that strain. Use
The following strains or those considered to be the inoculum suspensions within 24 hours after they
equivalent are used as the test microorganisms. have been prepared from the cultivations on agar plate
media or in liquid media. Store the inoculum suspen-
Escherichia coli ATCC 8739, NBRC 3972, KCTC sions in a refrigerator if it is not possible to inoculate
2571 them into the test specimen within 2 hours. Titrate the
Pseudomonas aeruginosa ATCC 9027, NBRC viable cell count of the inocula immediately before
13275, KCTC 2513 use, and then calculate the theoretical viable cell count
Staphylococcus aureus ATCC 6538, NBRC 13276, per mL (g) of the product present just after inoculation.
KCTC 1916, 1927 or 3881
Candida albicans ATCC 10231, NBRC 1594, 3. Test Procedure
JCM 2085, KCTC 7965 3.1. Category I products
Asperigillus niger ATCC 16404, NBRC 9455, Inject each of the cell suspensions aseptically into
KCTC 6196 or 6317 five containers containing the product and mix uni-
formly. When it is difficult to inject the cell suspen-
KP X 1873

sion into the container aseptically or the volume of the miscibility between the liquid medium and semisolid
product in each container is too small to be tested, ointments or oils in which test microorganisms were
transfer aseptically a sufficient volume of the product inoculated. Sometimes, these agents also serve to inac-
into each of alternative sterile containers, and mix the tivate or neutralize many of the most commonly used
inoculum. When the product is not sterile, incubate preservatives.
additional containers containing the uninoculated
product as controls and calculate their viable cell 4. Interpretation
counts (the viable counts of bacteria and those of Interpret the preservative efficacy of the product
yeasts and moulds). A sterile syringe, spatula or glass according to Table 1. When the results described in
rod may be used to mix the cell suspension uniformly Table 1 are obtained, the product examined is consid-
in the product. The volume of the suspension mixed in ered to be effectively preserved. There is a strong pos-
the product must not exceed 1/100 of the volume of sibility of massive microbial contamination having
the product. Generally, the cell suspension is inoculat- occurred when microorganisms other than the inocu-
ed and mixed so that the concentration of viable cells lated ones are found in the sterile product to be exam-
is 105 to 106 cells per mL or per gram of the product. ined, and caution is required in the test procedures
Incubate these inoculated containers at 20 C to 25 C and/or the control of the manufacturing process of the
with protection from light, and calculate the viable cell product. When the contamination level in a non-sterile
count of 1 mL or 1 g of the product taken at 0, 14 and product to be examined exceeds the microbial enu-
28 days subsequent to inoculation. Record any marked meration limit specified in “Microbial
changes (e.g., changes in color or the development of
a bad odor) when observed in the mixed samples dur- Table 1. Interpretation criteria by product category
ing this time. Such changes should be considered
when assessing the preservative efficacy of the prod- Interpretation criteria
Prod Microor-
uct concerned. Express sequential changes in the via- After 14 days After 28 days
cate ganisms
ble counts as percentages, with the count at the start of
the test taken as 100. Titration of the viable cell counts 0.1 % of in- Same or less
is based, in principle, on the Pour Plate Methods in Cat- Bacteria oculum count than level after
“Microbial Limit Test”. In this case, confirm whether egory or less 14 days
any antimicrobial substance is present in the test spec- IA Yeasts Same or less Same or less
imen. If a confirmed antimicrobial substance needs to /moulds than inoculum than inoculum
be eliminated, incorporate an effective inactivator of 1count
% of inocu- Same or less
count
the substance in the buffer solution or liquid medium Cat- Bacteria lum count or than level after
to be used for dilution of the test specimen, as well as egory less 14 days
in the agar plate count medium. However, it is neces- IB Yeasts Same or less Same or less
sary to confirm that the inactivator has no effect on the /moulds than inoculum than inoculum
growth of the microorganisms. When the occurrence count
10 % of inocu- count
Same or less
of the preservative or the product itself affects titration Bacteria lum count or than level after
Cat-
of the viable cell count and there is no suitable less 14 days
egory
inactivator available, calculate the viable cell counts
IC Yeasts Same or less Same or less
by the Membrane Filtration method in “Microbial
/moulds than inoculum than inoculum
Limit Test”.
Bacteria countor less
Same count
Same or less
3.2. Category II products
The procedures are the same as those described for Cat- than inoculum than inoculum
Category I products. However, special procedures and egory count count
considerations are required for both uniform disper- II Yeasts Same or less Same or less
sion of the test microorganism in the product and titra- /moulds than inoculum than inoculum
tion of viable cell counts in the samples. countPharmaceutical
Attributes of Non-sterile count
Products” in
For semisolid ointment bases, heat the sample to General Information, caution is also required in the
45 C to 50 C until it becomes oily, add the cell sus- test procedures and/or the control of the manufactur-
pension and disperse the inoculum uniformly with a ing process of the product.
sterile glass rod or spatula. Surfactants may also be
added to achieve uniform dispersion, but it is neces- 5. Culture media
sary to confirm that the surfactant added has no effect Culture media and buffer solutions used for Pre-
on survival or growth of the test microorganisms and servatives-Effectiveness Tests are described below.
that it does not potentiate the preservative efficacy of Other media may be used if they have similar nutritive
the product. For titration of the viable cell count, a ingredients and selective and growth-promoting prop-
surfactant or an emulsifier may be added to disperse erties for the microorganisms to be tested.
the product uniformly in the buffer solution or liquid Soybean Casein Digest Agar Medium
medium. For instance, sorbitan monooleate, Casein peptone 15 g
polysorbate 80 or lecithin may be added to improve Soybean peptone 5.0 g
1874 Monographs, Part II

Sodium chloride 5.0 g can be measured by a volumetric or continuous flow


Agar 15.0 g procedure.
Water 1000 mL
Mix all of the components and sterilize at 121 C Multi-Point Measurement
for 15 ~ 20 minutes in an autoclave. The medium pH When the gas is physically adsorbed by the pow-
after sterilization: 7.1 ~ 7.3. der sample, the following relationship (Brunauer,
Sabouraud  Glucose Agar Medium Emmett and Teller (BET) adsorption isotherm) holds
Peptone (animal tissue an casein) 10.0 g when the relative pressure (P/P0) is in the range of
Glucose 40.0 g 0.05 to 0.30 for pressure P of the adsorbate gas in
Agar 15.0 g equilibrium for the volume of gas adsorbed, Vα.
Water 1000 mL
Mix all of the components and sterilize at 121 C 1 (C  1) P 1
   (1)
for 15 ~ 20 minutes in an autoclave. The medium pH P Vm C P0 Vm C
after sterilization: 5.4 ~ 5.8. Va ( 0  1)
P
Glucose  Peptone (GP) Agar Medium
Glucose 20.0 g P: Patial vapour pressure of adsorbate gas in equilib-
Yeast extract 2.0 g rium with the surface at -195.8 ºC (b.p. of liquid
Magnesium sulfate heptahydrate 0.5 g nitrogen) (Pa)
Peptone 5.0 g P0: Saturated pressure of adsorbate gas (Pa)
Monobasic potassium phosphate 1.0 g Va: Volume of gas adsorbed at saturated temperature
Agar 15.0 g and pressure (STP) [0 ºC and atmospheric pressure
Water 1000 mL (1.013 × 105 Pa)] (mL)
Mix all of the components and sterilize at 121 C Vm: Volume of gas adsorbed at STP to produce ap-
for 15 ~ 20 minutes in an autoclave. The medium pH parent monolayer on the sample surface (mL)
after sterilization: 5.6 ~ 5.8. C: Dimensionless constant that is related to the en-
Potato  Dextrose Agar Medium thalpy of adsorption of adsorbate gas on the pow-
Potato extract 4.0 g der sample.
Glucose 20.0 g
Agar 15.0 g A value of Va is measured at each of not less than
Water 1000 mL 3 values of P/P0. Then the BET value, 1/[Va{P0/P-1}],
Mix all of the components and sterilize at 121 C is plotted against P/P0 according to equation (1). This
for 15 ~ 20 minutes in an autoclave. The medium pH plot should yield a straight line usually in the approx-
after sterilization: 5.4 ~ 5.8 imate relative pressure range 0.05 to 0.30. The data
0.1% Peptone Water are considered acceptable if the correlation coefficient,
Peptone 1.0 g r, of the linear regression is not less than 0.9975; that
Sodium chloride 8.0 g is, r2 is not less than 0.995. From the resulting linear
Water 1000 mL plot, the slope, which is equal to (C-1)/VmC, and the
Mix all of the components and sterilize at 121 C intercept, which is equal to 1/VmC, are evaluated by
for 15 ~ 20 minutes in an autoclave. The medium pH linear regression analysis. From these values, Vm is
after sterilization: 7.2 ~ 7.4. calculated as 1/(slope + intercept), while C is calculat-
ed as (slope/intercept) + 1. From the value of Vm so
determined, the specific surface area, S (m2/g) is cal-
12. Specific Surface Area culated by the equation:

Determination Method (V m Na)


S (2)
m  22400
The specific surface area determination method is
a method to determine specific surface area (the total N: Avogadro constant (6.022 × 1023 /mol)
surface area of powder per unit mass) of a pharmaceu- a: Effective cross-sectional area of one adsorbate
tical powder sample by using gas adsorption method. molecule (m2)
The specific surface area of a powder is determined by N2: 0.162 × 10-18
physical adsorption of a gas on the surface of the solid Kr: 0.195 × 10-18
and by calculating the amount of adsorbate gas corre- m: mass of test powder (g)
sponding to a monomolecular layer on the surface.
Physical adsorption results from relatively weak forc- Single-point measurement
es (van der Waals forces) between the adsorbate gas Normally, at least 3 measurements of Va each at
molecules and the adsorbent surface of the test powder. different values of P/P0 are required for the determina-
The determination is usually carried out at the temper- tion of specific surface area by the dynamic flow gas
ature of liquid nitrogen. The amount of gas adsorbed adsorption technique (Method I) or by volumetric gas
KP X 1875

adsorption (Method II). However, under certain cir- samples, other outgassing methods such as the desorp-
cumstances described below, it may be acceptable to tion-adsorption cycling method may be employed.
determine the specific surface area of a powder from a The standard technique is the adsorption of nitro-
single value of Va measured at a single value of P/P0 gen at liquid nitrogen temperature.
such as 0.30, using the following equation for calcu- For powders of low specific area (< 0.2 m2/g) the
lating Vm: proportion adsorbed is low. In such cases the use of
krypton at liquid nitrogen temperature is preferred
P because the low vapor pressure exerted by this gas
Vm  Va (1  ) (3)
greatly reduces error.
P0
All gases used must be free from moisture.
Accurately weigh a quantity of the test powder
The single-point method may be employed di- such that the total surface of the sample is at least 1 m2
rectly for a series of powder samples of a given mate- when the adsorbate is nitrogen and 0.5 m2 when the
rial for which the material constant C is much greater adsorbate is krypton. Lower quantities of sample may
than 1. Close similarity between the single-point val- be used after appropriate validation.
ues and multiple-point values suggests that 1/C ap- Adsorption of gas should be measured either by
proaches zero. The error on Vm can be reduced by us- Method I or Method II.
ing the multiple-point method to evaluate C for one of
the samples of the series on which the material con- Method I: the dynamic flow method
stant C is expected to be large. Then Vm is calculated In the dynamic flow method (see Fig. 1), the rec-
from the single value of Va measured at a single value ommended adsorbate gas is dry nitrogen or krypton,
of P/P0 by the equation: while helium is employed as a diluents gas, which is
not adsorbed under the recommended conditions. A
P  1 C  1 P  minimum of 3 mixtures of the appropriate adsorbate
Vm  Va  0  1    (4)
P  C C P0  with helium are required within the P/P0 range 0.05 to
0.30.
Sample preparation
Before the specific surface area of the sample can
be determined, it is necessary to remove gases that
may have become physically adsorbed onto the sur-
face during storage and handling. If outgassing is not
achieved, the specific surface area may be reduced or
may be variable because an intermediate area of the
surface is covered with molecules of the previously
adsorbed gases or vapors. The outgassing conditions
are critical for obtaining the required precision and
accuracy of specific surface area measurements on
pharmaceuticals because of the sensitivity of the sur-
face of the materials. The outgassing conditions must
be demonstrated to yield reproducible BET plots, a
A: Flow control valve
constant weight of test powder, and no detectable
B: Differential flow controller
physical or chemical changes in the test powder.
C: On-off valve
The outgassing conditions defined by the temper-
D: Gas inlet
ature, pressure and time should be so chosen that the
E: O ring seals
original surface of the solid is reproduced as closely as
F: Cold trap
possible.
G: Thermal equilibration tube
Outgassing of many substances is often achieved
H: Detector
by applying a vacuum, by purging the sample in a
I: Digital display
flowing stream of a non-reactive, dry gas, or by apply-
J: Calibration septum
ing a desorption-adsorption cycling method. In either
K: Sample cell
case, elevated temperatures are sometimes applied to
L: Self seals quick connection
increase the rate at which the contaminants leave the
M: Short path ballast
surface. Caution should be exercised when outgassing
N: Detector
powder samples using elevated temperatures to avoid
O: Path selection valve
affecting the nature of the surface and the integrity of
P: Long pass ballast
the sample.
Q: Flow meter
If heating is employed, the recommended tem-
R: Outgassing station
perature and time of outgassing are as low as possible
S: Diffusion baffle
to achieve reproducible measurement of specific sur-
T: Vent
face area in an acceptable time. For heating sensitive
1876 Monographs, Part II

Fig. 1. Schematic diagram of the dynamic flow Admit a small amount of dry nitrogen into the sample
method apparatus tube to prevent contamination of the clean surface of
sample, remove the sample tube, insert the stopper,
The gas detector-integrator should provide a sig- and weigh it. Calculate the weight of the sample. At-
nal that is approximately proportional to the volume of tach the sample tube to the volumetric apparatus. Cau-
the gas passing through it under defined conditions of tiously evacuate the sample down to the specified
temperature and pressure. For this purpose, a thermal pressure (e.g. between 2 Pa and 10 Pa).
conductivity detector with an electronic integrator is If the principle of operation of the instrument requires
one among various suitable types. A minimum of 3 the determination of the dead volume in the sample
data points within the recommended range of 0.05 to tube, this procedure is carried out at this point, fol-
0.30 for P/P0 is to be determined. lowed by evacuation of the sample. Raise a Dewar
A known mixture of the gases, usually nitrogen vessel containing liquid nitrogen up to a defined point
and helium, is passed through a thermal conductivity on the sample cell. Admit a sufficient volume of
cell and then to a recording potentiometer. adsorbate gas to give the lowest desired relative pres-
Immerse the sample cell in liquid nitrogen, then sure. Measure the volume adsorbed, Va. For multipoint
the sample adsorbs nitrogen from the mobile phase. measurements, repeat the measurement of Va at suc-
This unbalances the thermal conductivity cell, and a cessively higher P/P0 values. When nitrogen is used as
pulse is generated on a recorder chart. the adsorbate gas, P/P0 values of 0.10, 0.20, and 0.30
Remove from the coolant; this gives a desorption are often suitable.
peak equal in area and in the opposite direction to the
adsorption peak. Reference materials
Since this is better defined than the adsorption Periodically verify the functioning of the apparatus
peak, it is the one used for the determination. using appropriate reference materials of known sur-
To effect the calibration, inject a known quantity face area, such as α-alumina for specific surface area
of adsorbate into the system, sufficient to give a peak determination, which should have a specific surface
of similar magnitude to the desorption peak and obtain area similar to that the sample to be examined.
the proportion of gas volume per unit peak area.
Method II: the volumetric method
In the volumetric method (see Figure 2), the rec-
ommended adsorbate gas is nitrogen. And it is admit-
13. Terminal Sterilization and
ted into the evacuated space above the previously out- Sterilization Indicators
gassed powder sample to give a defined equilibrium
pressure, P, of the gas. The use of a diluent gas, such Sterilization is a process whereby the killing or
as helium, is therefore unnecessary, although helium removal of all forms of viable microorganisms in sub-
may be employed for other purposes, such as to meas- stances is accomplished. It is achieved by terminal
ure the dead volume. sterilization or a filtration method. For substances to
which terminal sterilization can be applied, an appro-
priate sterilization method should be selected in ac-
cordance with the properties of the product, including
the packaging, after full consideration of the ad-
vantages and disadvantages of each sterilization meth-
od, from the heat method, irradiation method and gas
method. After installation of the sterilizer (including
design and development of the sterilization process),
validation is required to confirm that the sterilization
process is properly performing its designed function,
under conditions of loading and unloading of the
product, on the basis of sufficient scientific evidence.
After the process has been validated and the steriliza-
tion of the product commenced, the process must be
A: Vacuum gauge controlled correctly, and qualification tests of the
B: Nitrogen reservoir equipment and procedures must be performed regular-
C: Helium reservoir ly. The bioburden per product, prior to terminal sterili-
D: Vapor pressure manometer zation, must be evaluated periodically or on the basis
E: Vacuum and air of batches. Refer to the ISO standard (ISO 11737-1)
F: Cold traps and vacuum pumps relevant to bioburden estimation. For a substance to
which terminal sterilization can be applied, generally
Fig. 2. Schematic diagram of the volumetric method use sterilization conditions such that a sterility assur-
apparatus ance level of less than 10-6 can be obtained. The prop-
KP X 1877

erty of the sterilization should be judged by employing Therefore, in routine sterilization process control,
an appropriate sterilization process control, with the it is required to monitor continuously the tem-
use of a suitable sterilization indicator, and if neces- perature and exposure time, and they should be
sary, based on the result of the sterility test. included in the specifications of the sterilizer.
The filtration procedure is used for the sterilization of 2.2 Irradiation method
a liquid product, to which terminal sterilization cannot Microorganisms are directly killed by ionizing radia-
be applied. Concerning the disinfection and/or sterili- tion, or by the heat generated by microwave radiation.
zation necessary for processing equipment and areas (i) Radiation method
of pharmaceutical products, and performing microbio- Ionizing radiations which may be used are gam-
logical tests specified in the monographs, see Disin- ma () rays emitted from a radioisotope such as 60C,
fection and Sterilization Methods. an electron beam and bremsstrahlung (X rays) gener-
ated from an electron accelerator. Although any pro-
1. Definitions cedure can be applied to thermally unstable products
The definitions of the terms used in this text are with no radioactivity residue, it is necessary to consid-
as follows. er the possibility of material degradation. Although a
Terminal sterilization: A process whereby a prod- 25 kGy dose is traditionally used as a sterilization
uct is sterilized in its final container or packaging, and dose, there are some ways to calculate the dose as
which permits the measurement and evaluation of follows: the bioburden of the substance to be sterilized
quantifiable microbial lethality. is measured and the sterilization dose is calculated
Product: A generic term used to describe raw ma- based on the mean bioburden and the standard re-
terials, intermediate products, and finished products, sistance distribution (Method I in ISO 11137), the
to be sterilized. dose is calculated from the fraction positive infor-
Bioburden: Numbers and types of viable micro- mation from a sterility test in which representative
organisms in a product to be sterilized. product samples are exposed to a substerilizing dose
Sterility assurance level (SAL): Probability of a (Method 2 in ISO 11137), or the dose is calculated
viable microorganism being present in a product unit based on the bioburden and D value of the most re-
after exposure to the proper sterilization process, ex- sistant microorganisms (Log method) (see 5-3). In the
pressed as 10-n. case of the radiation sterilization procedure, factors
Integrity test: A non-destructive test which is which may affect the sterilization include dose (ab-
used to predict the functional performance of a filter sorbed dose) and exposure time. Therefore, in  ray
instead of the microorganism challenge test. sterilization process control, it is required to determine
D value: The value which shows the exposure the dose (the absorbed dose) at appropriate intervals
time (decimal reduction time) or absorbed dose (deci- and to monitor continuously the exposure time in
mal reduction dose) required to cause a 1-logarithm or terms of the operation parameters (the conveyer speed
90 % reduction in the population of test microorgan- and the cycle time). The dose control mechanism
isms under stated exposure conditions. should be included in the specifications of the steriliz-
Sterilization indicator: Indicators used to monitor er. In the case of electron beam or bremsstrahlung
the sterilization process, as an index of sterility, in- irradiation, it is required to monitor the acceleration
cluding biological indicators (BI), chemical indicators voltage, the beam current and beam scanning width
(CI), dosimeters and the like. besides the above-mentioned items.
(ii) Microwave method
2. Sterilization Microorganisms are killed by the heat generated
2.1 Heat method by microwave radiation, usually at the frequency of
In the heat method, microorganisms are killed by heat- 2450 50 MHz. This method is applied to liquids or
ing. water-rich products in sealed containers. Since a glass
(i) Moist heat method or plastic container may be destroyed or deformed due
Microorganisms are killed in saturated steam un- to the rise of the inner pressure, the containers must be
der pressure. In this method, factors which may certified to be able to withstand the heat and the inner
affect the sterilization include temperature, steam pressure generated during microwave sterilization.
pressure and exposure time. Therefore, in routine Leakage of electromagnetic radiation must be at a
sterilization process control, it is required to sufficiently low level to cause no harm to humans and
monitor continuously the temperature, steam no interference with radio communications and the
pressure and exposure time, and they should be like. In this method, factors which may affect the steri-
included in the specifications of the sterilizer. lization include temperature, processing time and mi-
(ii) Dry heat method crowave output power. Therefore, in routine steriliza-
Microorganisms are killed in dry heated air. This tion process control, it is required to monitor continu-
method is usually conducted in a batch-type dry ously the temperature, time and the microwave output
heat sterilizer or a funnel-type dry heat sterilizer. power, and they should be included in the specifica-
In this method, factors which may affect the ster- tions of the sterilizer.
ilization include temperature and exposure time. 2.3 Gas Method
1878 Monographs, Part II

Ethylene oxide (EO) is widely used as a steriliza- Table 1. Typical examples of biological indicator
tion gas. Since EO gas has an explosive nature, a 10 – Representative micro-
Sterilization Strain name
30 % mixture with carbon dioxide is commonly used. organisms*
Also, as EO gas is a strong alkylating agent, it cannot ATCC 7953,
be applied to the products which are likely to react NBRC 13737,
with or absorb it. Furthermore, because EO gas is tox- JCM 9488,
ic, the residual concentration of EO gas and other sec- Moist heat Geobacillus KCTC 2107
ondary generated toxic gases in products sterilized method stearothermophilus ATCC 12980,
with EO gas must be reduced to less than the safe lev- NBRC 12550,
els thereof by means of aeration and the like before JCM 2501,
the product is shipped. In this method, factors which KCTC 1752
may affect the sterilization include temperature, gas ATCC 9372,
concentration (pressure), humidity and exposure time. Dry heat
Bacillus atrophaeus NBRC 13721
Therefore, in routine sterilization process control, it is method
KCTC 1022
required to monitor continuously the temperature, gas
ATCC 9372,
concentration (pressure), humidity and exposure time,
Gas method Bacillus atrophaeus NBRC 13721
and they should be included in the specifications of
KCTC 1022
the sterilizer.
* In addition to these microorganisms, other microor-
3. Filtration method ganisms with the greatest resistance to the sterilization
Microorganisms are removed by using a steriliz- procedure concerned, found in the bioburden, can be
ing filter made of an appropriate material. However, used as the biological indicator.
this method is not intended for microorganisms small-
er than bacteria, Generally, a sterilizing filter chal- 4.1.1 D value of BI
lenged with more than 107 microorganisms of a strain Methods for determination of the D value include
Brevundimonas diminuta (ATCC 1914, NBRC 14213, the survival curve method and the fraction negative
JCM 2428), cultured under the appropriate conditions, method (Stumbo, Murphy & Cochran method, Lim-
per square centimeter of effective filter area should ited Spearman-Karber method and the like). In using
provide a sterile effluent. In this method, factors marketed BIs, it is usually unnecessary to determine
which may affect the sterilization include pressure, the D value before use if the D value indicated on the
flow rate, filter unit characteristics and the like. In label has been determined by a standardized biological
routine filtration process control, it is required to per- indicator evaluation resistometer (BIER) under strictly
form integrity tests of the sterilizing filter after each prescribed conditions in accordance with ISO 11138-1.
filtration process (also prior to the filtration process, if It is acceptable that the D value indicated on the label
necessary). shows a scattering of not more than 30 seconds.
4.1.2 Setting up procedure of BI
4. Sterilization Indicators (i) In the case of dry materials
4.1 Biological Indicator (BI) A dry type BI is placed at predetermined cold
A BI is prepared from specific microorganisms spots in the product to be sterilized or a suitable prod-
resistant to the specified sterilization process and is uct showing an equivalent effect in the sterilization.
used to develop and/or validate a sterilization process. The BIs are usually primary packaged in the same way
The dry type BI is classified into two kinds. In one, as the product, including a secondary packaging, if
bacterial spores are added to a carrier such as filter applicable.
paper, glass or plastic and then the carrier are dried (ii) In the case of wet materials
and packaged. In the other, bacterial spore are added Spores are suspended as the BI in the same solu-
to representative units of the product to be sterilized or tion as the product or in an appropriate similar solu-
to simulated products. Packaging materials of the BI tion, and should be placed at cold spots in the sterilizer.
should show good heat penetration in dry heat sterili- 4.1.3 Culture conditions of BI
zation and good gas or steam penetration in ethylene Soybean casein digest medium is generally used.
oxide and moist heat sterilizations. It should be con- General culture conditions are at 55 ~ 60 ℃ for 7 days
firmed that any carrier does not affect the D value of in the case of G. stearothermophilus and at 30 ~ 35 ℃
the spores. In the case of a liquid product, the spores for 7 days in the case of B. atrophaeus.
may be suspended in the same solution as the product
or in a solution showing an equivalent effect in the 4.2 Chemical indicator (CI)
sterilization of biological indicator. However, when CI is an indicator which shows a color change of
the spores are suspended in liquid, it is necessary to a substance applied to a paper slip, etc. as a result of
ensure that the resistance characteristics of the spores physical and/or chemical change due to exposure to
are not affected due to germination. Typical examples heat, gas or radiation. The CI can be classified into
of biological indicator are shown in Table 1. three types. The first is employed to identify whether
or not sterilization has already been implemented, the
KP X 1879

second is employed to control the sterilization process extensive bioburden estimation is considered as the
(for example, its color changes after sterilization for a maximum bioburden count, and the sterilization time
sufficient time), and the third is the Bowie & Dick (or radiation dose) is calculated with the bioburden
type used to evaluate the effectiveness of air removal count based on an objective sterility assurance level.
during the pre-vacuum phase of the pre-vacuum steri- When this procedure is used, it is required to deter-
lization cycle. mine the resistance of the bioburden to the steriliza-
4.3 Dosimeter tion as well as the bioburden count in the product be-
In the radiation (-ray) method, the sterilization ing sterilized. If a more resistant microorganism than
effect depends on the absorbed radiation dose, so the the BI spore is found in the bioburden estimation, it
sterilization process control is mainly performed by should be used as the BI.
measuring the dose. A dosimeter is installed at a posi-
tion corresponding to the minimum dose region of an N0
exposed container or a position where the dose is in a Sterilization time(or radiation dose)  D  log( )
N
known relation to that in the above region. Measure-
ment should be done for each radiation batch. If there D : D value of the BI
are many containers in the same batch, dosimeters N: Sterility assurance level
should be employed so that more than one dosimeter N0: Maximum bioburden count in the product
is always installed at the effective radiation section of 5.4 Absolute bioburden method
the irradiation chamber. It should be noted that dosim- The sterilization conditions are determined by
eters may be affected by environmental conditions employing the D value of the most resistant microor-
(temperature, humidity, ultraviolet light, time to read- ganism found in the product or environment by the
ing, etc.) before and during irradiation. Practical do- resistant estimation and being based on the bioburden
simeters for –ray and bremsstrahlung sterilization count in the product. Generally, a count of mean
include the dyed polymethylmethacrylate dosimeter, bioburden added three times of its standard deviation
clear polymethymethacrylate dosimeter, ceric-cerous obtained through the extensive bioburden estimation is
sulfate dosimeter, alanine-EPR dosimeter and the like. employed as the bioburden count. When this proce-
A dosimeter for gamma radiation cannot generally be dure is used, it is required to make frequent counting
used for sterilization process control by the electron and resistance determination of microorganisms in
beam energy less than 3 MeV. Dosimeters for electron daily bioburden estimation
beam sterilization include the cellulose acetate dosim-
eter, radiochromic film dosimeter and the like. A prac-
tical dosimeter must be calibrated against an appropri-
ate national or international standard dosimetry system.

5. Determination of sterilization conditions using


microorganism as an indicator
Taking account of the characteristics upon the
sterilization concerned, bioburden, etc., of a product to
be sterilized, choose a suitable method from the fol-
lowings and determine the conditions.
5.1 Half-cycle method
In this method, a sterilization time of twice as
long as that required to inactivate all of 106 counts of
BI placed in the product is used, regardless of
bioburden count in the product being sterilized or the
resistance of the objective microorganisms to the steri-
lization.
5.2 Overkill method
In this method, a sterilization condition giving a
sterility assurance level of not more than 10-6 counts is
used, regardless of bioburden count in the product
being sterilized or the resistance of the objective mi-
croorganisms to the sterilization. Generally, a steriliza-
tion condition providing 12 logarithmic reduction
(12D) of known count of BI of more than 1.0 D value
is used.
5.3 Combination of BI and bioburden
Generally, a count of mean bioburden added
three times of its standard deviation obtained by an

You might also like