Dissolution Testing: A Seminar On
Dissolution Testing: A Seminar On
DISSOLUTION TESTING
Presented by
Sukanya Patil, Zubiya Shah, and Mushahid Shaikh
(M Pharm sem II )
Department of Pharmaceutics
H. K. College of Pharmacy
TABLE OF CONTENT:
ALTERNATIVE METHODS OF DISSOLUTION TESTING
CATEGORIES OF IVIVC:
• LEVEL A CORRELATION
• LEVEL B CORRELATION
• LEVEL C CORRELATION
ALTERNATIVE METHODS OF
DISSOLUTION TESTING
(1)ROTATING BOTTLE METHOD:
The rotating bottle method was suggested in NF-XIII (National Formulary) but has become less popular since.
The rotating bottle method was used mainly for controlled-release beads. For this purpose the dissolution
medium may be easily changed, such as from artificial gastric juice to artificial intestinal juice. The equipment
consists of a rotating rack that holds the sample drug products in bottles. The bottles are capped tightly and
rotated in a 37°C temperature bath. At various times, the samples are removed from the bottle, decanted
through a 40-mesh screen, and the residues are assayed. An equal volume of fresh medium is added to the
remaining drug residues within the bottles and the dissolution test is continued. A dissolution test with pH 1.2
medium for 1 hour, pH 2.5 medium for the next 1 hour, followed by pH 4.5 medium for 1.5 hours, pH 7.0
medium for 1.5 hours, and pH 7.5 medium for 2 hours was recommended to simulate the condition of the
gastrointestinal tract. The main disadvantage is that this procedure is manual and tedious.
(2) INTRINSIC DISSOLUTION METHOD:
Most methods for dissolution deal with a finished drug product. Sometimes a new drug or substance may be
tested for dissolution without the effect of excipients or the fabrication effect of processing. The dissolution of
a drug powder by maintaining a constant surface area is called intrinsic dissolution. Intrinsic dissolution is
usually expressed as mg/cm2/min. In one method, the Basket method is adapted to test dissolution of powder
by placing the powder in a disk attached with a clipper to the bottom of the basket.
The peristalsis method attempts to simulate the hydrodynamic conditions of the gastrointestinal tract in an in
vitro dissolution device. The apparatus consists of a rigid plastic cylindrical tubing fitted with a septum and
rubber stoppers at both ends. The dissolution chamber consists of a space between the septum and the lower
stopper. The apparatus is placed in a beaker containing the dissolution medium. The dissolution medium is
pumped with peristaltic action through the dosage form.
(4) DIFFUSION CELLS:
In order for the dissolution test to be performed properly, and give meaningful results, these three components
must interact together optimally, or the results can be misleading. The USP general chapter for dissolution
includes performance verification test (PVT), to assure the suitability of Apparatus 1 and 2 when used for testing
drug products. PVT requires chemical calibration with calibrator tablets that may be obtained from USP-NF. The
calibration tablets, either prednisone tablets for dissolution tests requiring disintegrating tablets or salicylic acid
as a standard for non disintegrating tablets, are used to qualify USP dissolution Apparatus 1 and Apparatus 2.
PVT is also useful to compare performance of different dissolution apparatus used in different laboratories.
Mechanical calibration is a critical component of the qualification of the dissolution apparatus. The FDA has
introduced a mechanical calibration approach that considers mechanical specifications of the instrument design
and its manufacture (FDA Guidance for Industry, January 2010). Instead of using a calibrator tablet, a
pharmaceutical manufacturer can use an appropriately rigorous method of mechanical calibration for dissolution
Apparatus 1 and 2.
(8) DISCRIMINATING DISSOLUTION TEST
The value of in vitro dissolution testing is its ability to characterize drug products and assist in decision making
including
(1) ensuring quality control through a linkage to batches used in pivotal clinical studies;
(2) information on batch-to-batch consistency; and
(3) guide in formulation development.
Dissolution testing is the only product test that truly measures the effect of formulation and physical properties
of the active pharmaceutical ingredient (API) on the rate of drug solubilization. In addition, under certain
circumstances (eg, presence of an adequate IVIVC) in vitro dissolution testing can serve as a surrogate for
bioequivalence studies to assess the impact of some pre and post approval changes. The dissolution testing
procedure should be discriminating to ensure its value. A discriminating method is the one that is appropriately
sensitive to manufacturing changes. A discriminating method is able to differentiate drug products manufactured
under target conditions versus drug products that are intentionally manufactured with meaningful variations (ie,
±10%–20% change) to the specification ranges of the most relevant material attributes and manufacturing
variables (eg, drug substance particle size, polymorphism, compression force, tablet hardness, etc). The choice of
experimental design to evaluate the most relevant variables will depend on the design of the dosage form, the
manufacturing process, and intrinsic properties of the API.
MEETING DISSOLUTION REQUIREMENTS
According to the Code of Federal Regulations (CFR), a drug product application should include the
specifications necessary to ensure the identity, strength, quality, purity, potency, and bioavailability of
the drug product, including, and acceptance criteria relating to, dissolution rate in the case of solid
dosage forms. For the selection of the dissolution acceptance criteria, the following points should be
considered:
1. The dissolution profile data from the pivotal clinical batches and primary (registration) stability
batches should be used for the setting of the dissolution acceptance criteria of your product (ie,
specification-sampling time point and specification value). A significant trend in the change in
dissolution profile during stability should be justified with dissolution profile comparisons and in
vivo data in those instances where the similarity testing fails.
2. Specifications should be established based on average in vitro dissolution data for each lot under
study, equivalent to USP Stage 2 testing (n = 12).
MEETING DISSOLUTION REQUIREMENTS
3. For immediate-release formulations, the last time point should be the time point where at least 80% of
drug has been released. If the maximum amount released is less than 80%, the last time point should be
the time when the plateau of the release profile has been reached. Percent release of less than 80% should
be justified with data (eg, sink conditions information).
4. For extended-release formulations, a minimum of three time points is recommended to set the
specifications. These time points should cover the early, middle, and late stages of the release profile. The
last time point should be the time point where at least 80% of drug has been released. If the maximum
amount released is less than 80%, the last time point should be the time when the plateau of the release
profile has been reached.
5. The dissolution acceptance criterion should be set in a way to ensure consistent performance from lot
to lot, and this criterion should not allow the release of any lots with dissolution profiles outside those
that were studied clinically.
The term Q means the amount of drug dissolved within a
given time period established in the drug product
specification table and is expressed as a per- centage of label
content.
However, if the dissolution of the drug is independent of the dissolution conditions (such as apparatus
agitation rate, pH, etc), then it is possible to establish such a correlation with only one formulation. The
establishment of a predictive IVIVC not only provides you with a better understanding of the release
properties of the drug product but also enables one to decrease the number of in vivo studies needed to
approve and maintain a drug product on the market resulting in an economic benefit as well as a decreased
regulatory burden. It also enables one to set clinically meaningful dissolution specifications based on the
predicted plasma concentration time profile.
A meaningful and predictive IVIVC is a correlation that is able to predict the Cmax and AUC within
20% (FDA guidance for industry, 1997b).
(1) Internal predictability refers to the ability to predict the pharmacokinetic profile of the
formulations that were used to develop the correlation;
(2) External predictability refers to the ability to detect the profile of a lot or formulation that was not
used to develop the IVIVC. In the United States and in Europe, a bioequivalence study can be waived
based on the IVIVC if the predicted mean AUC and Cmax of the test and reference do not differ from
each other by more than 20% (US IVIVC guidance for industry; EMA, August 2012).
CATEGORIES OF IVIVC
LEVEL A CORRELATION:
Level A correlation is the highest level of correlation and represents a point-to-point (1:1) relationship between
an in vitro dissolution and the in vivo input rate of the drug from the dosage form. Level A correlation
compares the percent (%) drug released versus percent (%) drug absorbed. Generally, the percentage of drug
absorbed may be calculated by the Wagner-Nelson or Loo–Riegelman procedures or by direct mathematical
deconvolution, a process of mathematical resolution of blood level into an input (absorption) and an output
(disposition) component.
The major advantage of a Level A correlation is that a point-to-point correlation is developed. All in vitro
dissolution data and all in vivo plasma drug concentration–time profile data are used. Once a Level A
correlation is established, an in vitro dissolution profile can serve as a surrogate for in vivo performance. A
change in manufacturing site, method of manufacture, raw material supplies, minor formulation modification,
and even product strength using the same formulation can be justified without the need for additional human
studies. Level A correlation enables the in vitro dissolution test to become meaningful and clinically relevant
quality control test that can predict in vivo drug product performance.
LEVEL B CORRELATION:
Level B correlation utilizes the principle of statistical moment in which the mean in vitro dissolution time is
compared to either the mean residence time (MRT)2 or the mean in vivo dissolution time (MDT). Level B
correlation uses all of the in vitro and in vivo data, but is not a point-to-point correlation. Different profiles can
give the same parameter values. The Level B correlation alone cannot justify formulation modification,
manufacturing site change, excipient source change, batch-to-batch quality, etc.
LEVEL C CORRELATION:
A Level C correlation is not a point-to-point correlation. A Level C correlation establishes a single-point
relationship between a dissolution parameter such as percent dissolved at a given time and a pharmacokinetic
parameter of interest such as AUC and Cmax. Level C correlation is useful for formulation selection and
development but has limited application. Multiple Level C correlation relates one or several pharmacokinetic
parameters of interest to the amount of drug dissolved at several time points of the dissolution profile. In general,
if one is able to develop a multiple Level C correlation, then it may be feasible to develop a Level A correlation.
Several examples of Level C correlation are given as followed.
Dissolution rate versus absorption rate