You are on page 1of 23

C H A P T E R

18
Dissolution
Vivian A. Gray1, Thomas W. Rosanske2
1
V. A. Gray Consulting, Inc., Hockessin, DE, United States; 2T.W. Rosanske Consulting, Overland Park,
KS, United States

O U T L I N E

18.1 Introduction 482 18.3.9 Method and equipment


parameters 489
18.2 The dissolution test 483
18.3.10 Manufacturing process 489
18.2.1 Apparatus 484
18.2.1.1 USP Apparatus 1 18.4 Method validation 489
(basket) 484 18.4.1 Linearity and range 490
18.2.1.2 USP Apparatus 2 18.4.2 Bias and accuracy 491
(paddle) 484 18.4.3 Intermediate precision and
18.2.1.3 USP Apparatus 3 reproducibility 491
(Reciprocating 18.4.4 Repeatability/method precision 492
Cylinder) 484 18.4.5 Specificity 492
18.2.1.4 USP Apparatus 4 18.4.6 Solution stability 493
(flow-through-cell) 485 18.4.7 Robustness 493
18.4.8 Other validation parameters 494
18.3 Method development 485
18.3.1 Solubility 486 18.5 Automation 494
18.3.2 Choice of media 487
18.6 Sources of error in dissolution testing 494
18.3.3 Sinkers 487
18.6.1 The dissolution apparatus 494
18.3.4 The “infinity point” 488
18.6.2 Dissolution vessels 495
18.3.5 Determination of discriminatory
18.6.3 Vibration and mechanical
power 488
aberration 495
18.3.6 Design of experiments and
18.6.4 Dissolved gases 495
quality by design concepts and
18.6.5 Standard solutions 496
variations 488
18.6.6 Method considerations 496
18.3.7 Drug substance properties 489
18.6.6.1 Sample introduction 496
18.3.8 Drug product properties 489
18.6.6.2 Dissolution media 496

Specification of Drug Substances and Products


https://doi.org/10.1016/B978-0-08-102824-7.00018-X 481 Copyright © 2020 Elsevier Ltd. All rights reserved.
482 18. Dissolution

18.6.6.3 Sinkers 497 18.9 Regulatory aspects and specifications 499


18.6.6.4 Filters 497 18.9.1 Immediate release drug products 500
18.6.6.5 Automation 497 18.9.2 Extended-release drug products 500
18.6.6.6 Cleaning 498 18.9.3 Generic drug products 501
18.6.6.7 Method transfer 498
18.10 Conclusions 501
18.7 Visual observations 498
18.11 Bibliography 501
18.8 Performance verification of
References 501
dissolution equipment 499

18.1 Introduction of formulation and manufacturing processes of


the dosage form. Dissolution is considered by
A dissolution test, as a release test, is required most regulatory agencies as a highly critical
for virtually all pharmaceutical products that are quality characteristic for most solid dosage
not true solutions. Dissolution testing monitors forms.
the rate at which a solid or semisolid pharmaceu- The regulatory agencies use the dissolution
tical dosage forms releases the active ingredi- test to provide a quality connection from a
ent(s) into a liquid medium at liquid/solid pivotal biobatch, if available, to the commercial-
interface, under standardized conditions of tem- ized product. For this reason, the dissolution test
perature, agitation, flow rate, volume, and me- development and validation are critical factors in
dia composition. Dissolution, or in vitro insuring that the test is robust and clinically rele-
release, of the drug substance from the product vant. Clinical relevance comes from developing
into a typically aqueous-based medium, is linked a test that provides understanding of the product
to the release of the drug into the body, making it release mechanism(s) and, in the highest form,
available for absorption, and then efficacy or an in vivoein vitro correlation (IVIVC). The
clinical outcome. Scientists have been con- acceptance criteria in the drug product (DP)
ducting dissolution studies for many years. specification or compendial monograph attrib-
However, it was not until 1970 that dissolution uted to a clinically relevant test are most impor-
testing was officially recognized as an indicator tant and useful.
of product quality when it was incorporated An important regulatory development has
into 12 monographs in the United States Phar- been the FDA Guidance for Industry, “Waiver
macopeia/National Formulary (USP/NF), USP of In Vivo Bioavailability and Bioequivalent Studies
XVIII/NF XIII.1 In the current US Pharmacopeial for Immediate Release Solid Oral Dosage Forms
edition, USP 42, nearly all monographs for solid Based on a Biopharmaceutics Classification System
dosage form include a dissolution test.2 (BCS).”4 This guidance is extremely significant
Dissolution is defined as a Category III test by because it affirms that under some circumstances
the USP, i.e., an “Analytical method for the determi- the dissolution testing can be used in lieu of
nation of performance characteristics .”3 Dissolu- bioavailability (BA) and/or bioequivalence (BE)
tion testing is primarily used in industry as a clinical studies. Whereas this guidance applies
quality control tool to monitor the consistency only to immediate release products, the general

IV. Specific tests: drug product


18.2 The dissolution test 483
principles therein may be also be applicable to considered. The analyst developing the dissolu-
controlled release (CR), or extended release tion test needs to know, for example, whether
(ER) under certain circumstances. The guidance the dosage form is a tablet, capsule, semisolid
defines the four classes of the biopharmaceutics (ointment or cream), or transdermal, and
classification system and the methods for deter- whether it is designed for immediate release or
mining the three aspectsdsolubility, perme- controlled release of the drug product. Of key
ation, and dissolution. importance is the potency of the dosage form
Development and validation of dissolution or the amount of drug to be delivered and the
methods require a good understanding of the rate at which the drug is to be delivered. This
theory of dissolution and the roles of the key pa- is related directly to the mathematical expression
rameters of the dissolution test. A complete of dissolution rate, which is defined by the
dissolution validation package would consider NoyeseWhitney equation:
at a minimum the dissolution apparatus used,
equipment qualification requirements, and any dW
¼ k1 SðCsat  Csol Þ (18.1)
appropriate governmental or regulatory guide- dt
lines. Therefore, it is important to address these where
issues here in the context of dissolution test
validation. dW/dt ¼ the dissolution rate
k1 ¼ the dissolution rate constant
Csat ¼ the concentration of a saturated
solution
18.2 The dissolution test
Csol ¼ the concentration of the solution at any
In order for a dissolution test to demonstrate given time
the unique dissolution characteristics of the S ¼ the surface area of the solid
dosage form, the dissolution procedure should In vivo, the gastrointestinal tract acts as a nat-
be based on the physical and chemical proper- ural sink, provided the drug is rapidly absorbed
ties of the drug substance as well as the dosage as it dissolves. In vitro, sink conditions are simu-
form design. Some of the physicalechemical lated by using either a large volume of dissolu-
properties of the drug substance and media tion medium or by replenishing the medium
which influence the dissolution characteristics with fresh solvent at a specific rate. By keeping
are the volume of dissolution medium at least three
• solubility in water and other appropriate times greater than the saturation volume, sink
solvents conditions are approximated. When sink condi-
• ionization constants tions are achieved, Csat >> Csol, and Eq. (18.1)
• solution stability simplifies to
• particle size/surface area dW
• crystal form ¼ k2 S (18.2)
dt
• common ion effects
where
• ionic strength
• buffer effects k2 ¼ a dissolution constant ¼ k1Csat
• octanol/water partition coefficients
In this case (Eq. 18.2), the dissolution rate is
• effect of temperature on solubility
characteristic of the release of active ingredient
Once the drug substance properties have been from the dosage form rather than the solubility
determined, the actual dosage form needs to be in the dissolution medium. Therefore, sink

IV. Specific tests: drug product


484 18. Dissolution

conditions are one of the main experimental pa- 18.2.1.2 USP Apparatus 2 (paddle)
rameters to be controlled in dissolution testing. The USP Apparatus 2 is currently the most
Sink conditions can be achieved by the appro- frequently used apparatus for the dissolution
priate selection of the dissolution apparatus testing of solid dosage forms. The dissolution
and dissolution medium. vessel used with this apparatus is the same as
for the USP Apparatus 1. However, the basket
assembly is replaced by a paddle of specified di-
18.2.1 Apparatus mensions as shown in Fig. 18.2. With this appa-
ratus, the dosage form is dropped directly into
The USP General Chapter <711> Dissolution5 the vessel containing the dissolution medium
describes several apparatus types used in disso- and allowed to sink to the bottom; the paddle
lution testing. Other compendia such as the is then rotated at a specified speed but can be
Pharmacopoeia of Japan (JP),6 the British Phar- immersed in the vessel when the dosage form
macopoeia (BP),7 and the European Pharmaco- is dropped as long as the rotation device is
poeia (Ph. Eur.)8 all include the Apparatus 1 switched off; this is the common industry prac-
and 2 dissolution equipment as described in tice. The USP specifies placing the dosage form
the USP. The USP Apparatus 1 (basket) and in the apparatus and immediately operating at
Apparatus 2 (paddle) are by far the most a specified rotational speed. The USP does not
frequently used for immediate release and state whether the paddle can or cannot be
most extended-release dosage forms. The USP immersed prior to addition of the dosage form,
Apparatus 1, 2, 3, and 4 are described in detail only that the rotation be started after the dosage
below. The USP Apparatus 3, 4, 5, 6, and 7 are form has been added. However, experience has
used less frequently than Apparatus 1 and 2, shown that care should be taken to ensure that
but are being used more frequently for charac- the dosage form sinks to the bottom of the vessel
terizing the release of the active ingredient and does not stick to the sides, which can result
from novel dosage forms.9 Descriptions of USP in different dissolution profiles. Dosage units
Apparatus 5, 6, and 7 are found in the USP Gen- that float should be weighed down with a sinker
eral Chapter <724> Drug Release10; these are (see Section 18.3.3. Sinkers) or tested with USP
devoted to transdermal patches. In vitro release Apparatus 1.
of transdermals is described in more detail in
Chapter 19. 18.2.1.3 USP Apparatus 3 (Reciprocating
Cylinder)
The Reciprocating Cylinder, shown in
18.2.1.1 USP Apparatus 1 (basket) Fig. 18.3, is often used as a research tool where
The USP Apparatus 1, shown schematically in a change of pH in the media during the test is
Fig. 18.1, consists of a covered vessel of specified necessary, for example, when testing enteric
shape and dimensions and capacity of 1000 mL, coated tablets. The dosage unit can be moved
a metallic shaft one end of which attaches to a from row to row, with the vessels in each row
motor, and a cylindrical metallic mesh basket containing media of different pH or composi-
that attaches to the opposite end of the shaft. tion. The equipment has a special configuration
The dosage form is placed inside of the basket for beaded products; the beads are contained
and the basket assembly is immersed in the by the screens in the upper and lower parts of
dissolution vessel containing a minimal volume the cell, yet the reciprocating motion allows for
of dissolution medium and rotated at a specified good mixing. The typical media volume is
speed. 200e250 mL.

IV. Specific tests: drug product


18.3 Method development 485

FIGURE 18.1 USP Apparatus 1, basket. Reproduced with permission from The United States Pharmacopeial Convention (USP)
©2017.

18.2.1.4 USP Apparatus 4 (flow-through-cell) 18.3 Method development


In USP Apparatus 4, the drug product is
positioned in a cell through which the disso- The reader is referred to a number of seminal
lution medium flows constantly, dissolving articles describing the development of dissolu-
the drug dosage unit. The liquid passes tion methods in the literature.11e15 There is also
through a filter at the top of the cell and is a comprehensive chapter in the USP, General
then collected in a reservoir. Because of the Chapter <1092>, titled USP General Informational
constant flow of media, an extended-release Chapter, The Dissolution Procedure: Development
product matrix can be monitored, or a and Validation, which is an excellent resource
poorly soluble product can continually be for method development and validation.16 The
in a sink environment. A diagram of a ICH Harmonized Tripartite Guideline Q2A:
unique flow-through-cell is shown in Text on Validation of Analytical Procedures is also
Fig. 18.4. a useful resource.17

IV. Specific tests: drug product


486 18. Dissolution

FIGURE 18.2 USP Apparatus 2, paddle. Reproduced with


permission from The United States Pharmacopeial Convention
(USP) ©2017.

18.3.1 Solubility
The solubility of the drug substance is an
important, though not the only, driving force
in the dissolution test (Eqs. 18.1 and 18.2). It
is, however, important first to know the solubi-
lity in three physiologically relevant media: FIGURE 18.3 USP Apparatus 3, Reciprocating Cylinder.
0.1 M HCl, 4.5 pH buffer, and 6.8 pH buffer. Reproduced with permission from The United States Pharmaco-
peial convention (USP) ©2017.
Therefore, the analyst should be familiar with
the BCS classification system4 and characterize
the drug according to the criteria provided substance in the formulation becomes the focus
(solubility and permeability). Poorly soluble of the method development. Proteolytic en-
drugs may require using alternative media, zymes may also be needed to dissolve pellicles
typically ones containing surfactants. Following formed during the dissolution testing of gelatin
an understanding the solubility of the drug sub- capsules, which can impede the release of the
stance, the dissolution rate of the drug drug.

IV. Specific tests: drug product


18.3 Method development 487
0.1 M HCl. For an extended-release product,
typically a pH 6.8 buffer is used because it
most resembles the pH of the small intestine.
For a delayed release formulation, the medium
typically consists of an acidic stage followed by
a pH 6.8 buffer stage.5 For poorly soluble drugs
where a solubility enhancer may be necessary,
there are a range of surfactants that may be suit-
able. The most commonly used surfactants are
sodium lauryl sulfate (SLS) and polysorbate so-
lutions, used at concentrations of 0.5%e2%.
The chosen concentration of surfactant must be
justified scientifically to be used in a regulatory
test.

18.3.3 Sinkers
Dosage forms that tend to float or move
around during the course of a dissolution test
can result in variability and bias. Sinkers are
often used to hold such dosage forms in place
during a dissolution test. There are many types
of sinkers that may be used in dissolution
testing, including custom-made sinkers, and
commercially manufactured sinkers. However,
it is important that the construction of the sinker
is well-defined because it can contribute to vari-
ability of the results. Therefore, when sinkers
are necessary, detailed descriptions and an
explanation of why a sinker is necessary must
be stated in the method. When comparing
different sinkers (or sinker vs. no sinker), tests
FIGURE 18.4 USP Apparatus 4, flow-through-cell.
must be run concurrently with each sinker.
Reproduced with permission from The United States Pharmaco- Each sinker type must be evaluated based on
peial Convention (USP) ©2017. its ability to maintain the dosage at the bottom
of the vessel without inhibiting drug release.
18.3.2 Choice of media A comparison of the different sinker types (or
no sinker) is performed using the same criteria
The evaluation of dissolution media starts for intermediate precision described in USP
with the medium or media in which the drug General Chapter <1092>.16 The variability at
is most readily soluble. The type of formulation all time points should be evaluated when the
then is considered. For fast dissolving drugs, method calls for the generation of a dissolution
the current FDA Guidance18 recommends profile rather than a single point determination.

IV. Specific tests: drug product


488 18. Dissolution

When transferring a method, the sinkers must profiles of formulations that are intentionally
be duplicated as closely as possible in any sub- manufactured with deliberate, meaningful varia-
sequent testing facility. tions of the most relevant critical manufacturing
parameters (e.g., 10%e20% change to the
ranges of selected parameters).19,20
18.3.4 The “infinity point” In the past, dissolution tests have generally
been conducted at a single time point resulting
The final time point selected for a dissolution
in fast dissolution rates. However, unless the
test does not necessarily correspond to complete
product is a very rapidly dissolving BCS Class
dissolution of drug. However, it is important to
1 or BCS Class 3, a slower profile test is typically
establish that, at some point, or under some con-
needed to give pertinent information about any
ditions, all of the drug material can be accounted
change in the product. Regulatory agencies pre-
for. This can be particularly useful when investi-
fer a method that is clinically relevant, that is,
gating out-of-specification results obtained by
one that shows an IVIVC or certain discrimina-
Assay or Uniformity of Dosage Units (see Chap-
tory power. Since clinically relevant specifica-
ter 11 for further discussion). Performing an “in-
tions are the becoming a requirement for the
finity point,” or fast-stir, test in the early product
characterization and regulation of drug prod-
development phases or routinely on samples in
ucts, laboratories are putting more resources
testing can give an indication of the recovery/ac-
into method development and the establishment
curacy, assuming that the drug is present at or
of an IVIVC or at least some relationship be-
around 100% of the label claim in the dosage
tween dissolution and in vivo performance.
unit. To obtain an infinity point, during the
normal test after the last time point is pulled,
the paddle or basket speed is increased to at least
150 rpm for 30e60 min without stopping the 18.3.6 Design of experiments and quality
test, after which time a further sample is taken. by design concepts and variations
Although there is no requirement for 100%
Development of a dissolution method should
dissolution in the profile, the infinity point can
be a team effort. Formulators provide informa-
provide supportive data when compared to the
tion on the critical quality attributes (CQAs)
content uniformity data. The infinity point data
and provide the prototype dosage forms that
for all six vessels can be compared to the Assay
reflect important formulation and processes var-
and the Uniformity of Dosage Units (weight uni-
iables studied by DoE studies. The principles of
formity or content uniformity) values in terms of
quality by design (QbD) can also be applied to
the mean and the variability. This may also pro-
assessment of dissolution methods. The DoE
vide useful information as to any artifacts from
studies may be useful to establish the critical
the dissolution method and/or the dosage
factors that influence the dissolution rate. The
form interacting with the media.
DoE can evaluate the dissolution operational
parameters21,22 or the product formulation
release attributes.23,24 They may provide under-
18.3.5 Determination of discriminatory
standing of the release mechanisms and deter-
power mine if the dissolution method can show
The regulatory agencies often ask sponsors to change to the CQAs. As an application of a
support the discriminatory ability of the selected QbD approach to dissolution, Li, et al. were
dissolution method. This discriminatory ability able to demonstrate the ability to predict changes
may be demonstrated by comparing dissolution in drug dissolution rate over time as a function of

IV. Specific tests: drug product


18.4 Method validation 489
temperature and humidity for two active ingre- video recording of the dissolution experiments
dients in an immediate release dosage form can also be particularly useful in this regard).
using ASAPprimeÒ, an accelerated stability Contributions to variability in the dissolution
assessment program.25 Such applications could results from the formulation can arise from poor
be useful in setting specifications to cover the uniformity of dosage units (either weight vari-
shelf life of a product. More detailed information ability or content variability), and reactions in
on DoE is found in Chapters 2 and 4. the dissolution solution leading to drug degra-
dation in situ. Film coating may cause sticking
of the dosage form to the vessel walls, thereby
18.3.7 Drug substance properties changing the hydrodynamics of the test. Upon
aging, gelatin capsule shells are known to form
Knowledge of drug substance properties, espe-
pellicles. Tablets may become harder or softer
cially the pH-solubility profile and the effects of
over time, depending upon the excipients and
surfactant, is essential. The effects of excipients
interaction with moisture, which may affect
in the formulation on the solubility of the drug
disintegration and hence the dissolution rate.
should also be well-understood. One needs to
anticipate precipitation of the drug as the pH
changes in the dissolution media, or as the 18.3.9 Method and equipment
amount of drug in solution increases. When the parameters
drug substance is poorly soluble, complete disso-
The final dissolution method used for routine
lution in the standard may not be possible
analysis should reflect the optimum choice of
without long sonication or mixing; therefore, it
medium, apparatus, and the sampling time
is common practice to use a small amount of
points. The apparatus can be operated using
alcohol to completely dissolve the drug substance
various speeds (typically 50e100 rpm with
in the standard solution. There is a limit of 5%
Apparatus 1 and 2), dip rate (Apparatus 3),
alcohol in the final solution so that the final mea-
and flow rate (Apparatus 4). The sampling
surement is not adversely affected.
time points may vary from 5 min with a rapidly
disintegrating tablet or suspension to many
hours with extended-release products.
18.3.8 Drug product properties
Assuming the drug product is manufactured
consistently, highly variable dissolution results 18.3.10 Manufacturing process
may indicate that the dissolution method is not The various processes involved in the manu-
robust, which can result in difficulty identifying facture of the dosage form should be investigated
trends and effects of formulation changes. There to determine which factors will affect the release
are two major causal factors that influence vari- mechanism. The factors most likely to affect the
ability: mechanical and formulatory. Mechanical dissolution of tablets are compression force,
contributions to variability can arise from the blending time, lubrication time, and temperature.
dissolution method conditions chosen, such as
incomplete deaeration and inappropriate paddle
speed. Careful visual observation of the dissolu- 18.4 Method validation
tion process in the vessel can often help identify
whether mechanical conditions, such as rota- As mentioned above, the USP General Infor-
tional speed, need to be altered (visual and/or mational Chapter <1092>16 is an excellent

IV. Specific tests: drug product


490 18. Dissolution

resource for dissolution method validation. Vali- the drug and can be characteristics of the drug
dation of a dissolution test method consists of substance, the formulation, the manufacturing
two parts. The first part, and the part that will process, or stability.
be given the most emphasis in this section, is Once the appropriate dissolution conditions
the validation of the dissolution test method, have been established, the analytical method
that is, the actual dissolution run and the should be suitably validated. The validation pa-
removal of the samples for analysis. The second rameters may vary depending on the intended
part is the analytical method used to determine use, but will typically include, at a minimum,
the concentration of the drug, which is usually linearity and range, accuracy, precision, speci-
UV spectrophotometry or HPLC with UV ficity, solution stability, and robustness. Each
detection. of these analytical parameters is discussed in
There are numerous aspects of the dissolu- detail elsewhere in this book in various con-
tion procedure that require validation and texts, and the general principles are the same
there are different levels of validation depend- for dissolution. This section will discuss the
ing on the phase of development. In early validation of parameters unique to dissolution
development, filtration, deaeration, linearity, testing. All dissolution testing must be per-
precision, solution stability, selectivity, and ac- formed on a qualified dissolution apparatus
curacy/recovery should be considered as part meeting the specified mechanical and perfor-
of the validation. During later development mance standards and with qualified associated
(Phase III and beyond) full validation studies, analytical instrumentation for the determina-
to include intermediate precision, automation tion of sample concentrations.
and robustness are added. This chapter focuses
on validation of methods used in later stages of
development, when full method validation is
necessary.
18.4.1 Linearity and range
It is presumed that full validation will be con- Linearity and range demonstrate the ability of
ducted on the final dissolution test method the dissolution method to obtain test results over
established for a new drug application or a mar- the range of expected concentrations that are pro-
keting authorization application. The ideal portional to the concentration of the analyte in
dissolution method should exhibit a low vari- the sample. Analytical detector linearity should
ability in the dissolution results, the solutions be established over the entire range of concentra-
should be stable in the dissolution media, and tions expected during the procedure. For imme-
there should be an established dissolution pro- diate release formulations, a concentration
file, using a minimum of three time points. The range of at least 50% of the lowest concentration
dissolution profile should be gradual with at expected in the dissolution vessel to 120% of the
least two points at or below 85% of the active dis- highest concentration is sufficient. For extended-
solved to satisfy the rules for an f2 analysis.26 The release products, the concentration range should
f2, or similarity factor, is a critical tool for demon- extend from approximately 10%e120% of that
strating bioequivalence, although it is not expected from dissolution of the entire dose. If
needed for BCS Class I drugs where the product an extended-release product is formulated in
dissolves rapidly, e.g., 85% in 15 min within the multiple strengths, the detector linearity should
relevant range of pH. The dissolution test be confirmed from 10% of the lowest strength
method should be discriminating and capable concentration to 120% of the highest strength
of detecting changes in the product CQAs, as concentration. When justified, it may be accept-
these attributes would influence the release of able to use different dissolution conditions to

IV. Specific tests: drug product


18.4 Method validation 491
cover a product where there is a large range of in situ or in a flask with media heated to 37 C.
dosage strengths. For fixed dose combination The drug must be fully dissolved before an ac-
products, it may be acceptable to use different curacy aliquot is taken. The order of addition
dissolution methods for the analysis of the indi- of the drug substance to the media, either before
vidual drugs provided suitable justification is or after the excipients, may be critical. For
provided.25 Typically, the concentration range example, some drugs need to be added to the
is divided into five evenly spaced concentrations. media without the excipients to wet properly,
All samples should be heated to 37 C (or the or vice versa where the excipients need to mix
specified dissolution temperature). The tempera- first before the drug substance is introduced.
ture is especially important if the samples are For poorly soluble drugs, the mixing process
poorly soluble and/or near sink conditions in may take some time before the drug is fully dis-
the vessel. solved. In some cases, drug powder cannot be
Solutions of drug substance used in deter- added directly, and addition of a diluted stock
mining linearity and range can be prepared solution is appropriate: for example, when the
with placebo to show if the sample solution is amount of drug required is too small to weigh
reflective of a real dissolution sample. Whenever accurately or the drug is electrostatic. The
possible, all solutions should be made from a generally acceptable measured recovery is 95%
common stock, using the dissolution media as e105% of the amount added. Low recoveries
the diluting solvent. As mentioned earlier, may be due to excipient interference27 or incom-
alcohol may be used to enhance the solubility plete dissolution.
of the poorly soluble drug. No more than 5%
organic is allowed in the final solution. Linearity
is generally considered acceptable if the linear
18.4.3 Intermediate precision and
regression correlation coefficient is  0.997, and
the y-intercept not significantly different from
reproducibility
zero at the 95% confidence limit. Intermediate precision evaluates the effects of
random events or variations in the laboratory on
the precision of the dissolution method. If
possible, it is advisable to change the environ-
18.4.2 Bias and accuracy
mental conditions in the laboratory to the extent
The bias of a method is a measure of the possible to anticipate potential issues during
agreement between the measured value and method transfer to other laboratories. Many
the true value. The accuracy of the dissolution times, method transfer studies are not successful
method, which is determined from the recovery because insufficient emphasis was placed on the
of drug spiked into the dissolution medium, is a intermediate precision study to rigorously chal-
measure of the agreement between measured lenge the method to intra-laboratory and inter-
value and a reference (or standard) value. In laboratory variations. Intermediate precision
general, accuracy is determined by preparing should be evaluated at all time points of the
multiple samples containing the drug and dissolution profile using the drug product for
other constituents present in the dosage form evaluation.
(e.g., excipients, coating materials) ranging in The dissolution profiles on the same drug
concentrations from below or at the expected product should be determined by at least two
concentration of the lowest profile point to different analysts, with each analyst individu-
above the highest concentration upon full ally preparing standard solutions and media
release. The samples can be prepared either (with different lots of reagents, if available).

IV. Specific tests: drug product


492 18. Dissolution

The analysts should use different dissolution 18.4.4 Repeatability/method precision


baths, different HPLC systems (including col-
umns and mobile phases), and the tests should A third form of precision, repeatability, or
be performed on different days. Each analyst method precision, is determined by replicate
should test and analyze 12 dosage unit samples measurements of standard and sample solu-
from the same lot. The mean values and the tions prepared at five concentrations by a single
relative standard deviations at each time point analyst on a single instrument. To show the
of the dissolution test between each analyst precision of the sample solution, the DS may
are compared. The results are generally consid- be prepared in a solution with placebo as a sub-
ered acceptable if the absolute mean difference stitute. It is important that the drug be
does not exceed 10% at time points when less completely in solution for this evaluation and
than 85% of the drug is dissolved and does preparation of a standard solution may be facil-
not exceed 5% for time points equal to or above itated by first dissolving the drug in small
85% dissolved drug.16 The 10% acceptance amount of alcohol if necessary. However, the
criteria may appear generous, but high vari- alcohol content in the final solution should
ability of some products, especially at the earlier not exceed 5% when the samples are measured
time points, is well established because it is sen- by UV spectrophotometry to avoid shifts in the
sitive to variability in tablet disintegration as absorption spectrum. General acceptance
well as dissolution. However, if variability of in- criteria for repeatability for HPLC procedures
dividual values is low and a 10% difference in are <1% RSD and for UV analysis <2% RSD.
means obtained, it may be prudent to look for Higher RSDs may be acceptable when working
causes of the bias. It is also useful to look for in- with low concentrations.
dividual values that appear to be significantly
outside the values for the other samples. Box-
plots and quantitative statistical tests for differ- 18.4.5 Specificity
ences between the two analysts should also be Specificity is the ability of the method to
considered. It is important to note the same determine the analyte of interest in the presence
acceptance criteria should be used for method potential interferences. Whereas it is not an abso-
validation and method transfer. Method repro- lute requirement that the analytical procedure
ducibility is a measure of inter-laboratory vari- used for the analysis of dissolution samples, it
ability and is best measured during method is convenient to use the same stability indicating
transfer. Inclusion of the receiving laboratory HPLC method used for Assay (see Chapters
in the method validation studies and training 9e11) because the validation of the Assay
of the analysts at the receiving laboratory can method can incorporate experiments that simul-
significantly improve the chances of successful taneously demonstrate the dissolution method is
transfer of dissolution methods. Even seem- fit for purpose. UV spectrophotometry is simpler
ingly simple steps, such as how the tablets are and faster than HPLC; however, the absence of
added to the dissolution vessels, can affect the interference from excipients, inks, dyes, and
test results. For example, a difference in capsule shells should be demonstrated. Interfer-
dissolution results may be observed if one ence from impurities related to the drug can be
analyst adds the tablets while the paddles are ignored provided they do not exceed 2% of the
rotating and the other analyst adds the tablets drug concentration in the sample.
before rotation of the paddles is started (the cor- If UV spectrophotometry is used for the anal-
rect procedure). ysis of dissolution samples, it is good practice to

IV. Specific tests: drug product


18.4 Method validation 493
demonstrate by HPLC that the drug does not may be necessary for very unstable drugs. If stor-
degrade significantly during the dissolution age under refrigerated or frozen conditions is
experiment producing degradation products necessary, the samples must be allowed to
with different UV spectra. The USP allows for a come to room temperature and inspected very
correction for the capsule shell interference pro- carefully for particulate matter to ensure that
vided the interference does not exceed 25% of any precipitated materials are redissolved before
the labeled drug concentration, in which case analysis.
the analytical method should be modified to
reduce the interference.5
18.4.7 Robustness
The robustness is the ability of the method to
18.4.6 Solution stability resist small changes in instrument settings and
Solution stability is the determination of sta- the experimental conditions. As discussed in
bility of sample and standard solutions over Chapter 10, the preferred approach to robustness
time under normal laboratory conditions. The testing in dissolution is by a statistical design of
standard solution and the sample solution are experiments (DoEs) approach, in which rela-
stored under representative conditions over a tively large changes to the instrument settings
reasonable period of time and the results are (notably stir rate, filters and temperature, flow
compared with those obtained with freshly pre- rate of the sampling device), composition of the
pared standard solutions. The standard solution dissolution medium (pH, volume, buffer concen-
should be at expected profile point concentra- tration, surfactant concentration, aeration), as
tions not exceeding 120% of the dosage form dis- well as sample position in the apparatus, and
solved. Sample solutions should also be the design of the sinkers (if applicable), are
compared in the same manner to a freshly pre- made in a systematic fashion to define the design
pared sample; a placebo plus drug solution space of the method. Mathematical treatment of
could be used instead of an actual sample. The the experimental data allows for the prediction
acceptable range for standard and sample stabil- of the control space within which smaller
ity samples is 98.0%e102.0% of the freshly pre- changes to the instrument settings and the
pared solutions (note: both increases and composition of the dissolution media will not
decreases in concentration can occur due to sol- significantly affect the rate of dissolution. The
vent evaporation or chemical degradation, control space should never be more restrictive
respectively).16 An expiration date and storage than the specifications of the instrumentation
conditions should be established based on the re- or the total errors of the analytical measurements
sults of the experiments, ideally at least 8 days, (see Chapters 2 and 9 for a more detailed expla-
which would allow storage of the standard and nation of total analytical errors). Thorough eval-
sample solutions for up to 1 week at room tem- uation of the robustness of the dissolution
perature. The sample and standard solutions method is essential for successful method trans-
should be tested at regular intervals over the fer and to ensure that the performance of the
preferred expiration period in the event that method is maintained over long periods of
shorter storage times are necessary. It is also routine application. If the analysis of dissolution
good practice to store the solutions in the refrig- samples is performed by HPLC, typically using
erator in the event that the drug is not suffi- the Assay procedure, the nature of the dissolu-
ciently stable for room temperature storage. tion samples should be taken into account dur-
Very occasionally, storage of frozen samples ing validation.

IV. Specific tests: drug product


494 18. Dissolution

18.4.8 Other validation parameters pulling the sample from the vessel simulta-
neously by manual and automated sampling
Other aspects of validation may include methods for each time point. Note that the
carryover of residual drug, effect of an in- correction for the volume withdrawn from the
residence probe (e.g., auto-sipper or fiber optic medium is doubled in the latter case.
probe), adsorption of drug onto the vessel,
tubing and filters, and cleaning and/or rinse cy-
cles. Evaluation of the filter generally includes 18.6 Sources of error in dissolution testing
preparation of a suitable standard solution
(lowest and highest profile concentrations are Several factors can contribute to errors and
recommended) and a completely dissolved bias in a dissolution test, arising from the equip-
sample solution. For the standard solutions, re- ment (apparatus and dissolution vessels), dis-
sults of the filtered solution (appropriate solved gases, standard solutions, and reference
discard volumes should be determined before- standards, vibration and other mechanical aber-
hand) are compared to those of the unfiltered rations. Critical factors related to running the
standard. For the filtered sample solution, re- method that are not well documented in the
sults should be compared to a fully dissolved written procedure can be a common cause of
and centrifuged sample solution. The accept- failed method transfers.
able range for standard and sample filtration ef-
ficiency is generally between 98% and 102% of
the unfiltered standards solutions and unfil- 18.6.1 The dissolution apparatus
tered but centrifuged sample solution.
USP Apparatus 1 and 2 can be sources of er-
ror if not closely inspected before using. Obvi-
ously, dimensions should be as specified.5 In
18.5 Automation cases of both baskets and paddles, shafts must
be straight and true. The paddles are sometimes
Automated sample collection has become partially coated with Teflon. This coating can
much more widely used in recent years. If peel and partially shed from the paddle,
possible, when conducting a validation of an causing flow disturbance of hydrodynamics
automated method, there should be a compari- within the vessel. Paddles can rust and become
son to a manual sampling method for the same nicked or dented; this can adversely affect
dissolution conditions. All profile time points dissolution hydrodynamics and also be a source
should be evaluated. This validation can be of contamination. Thorough cleaning of the
done in one of two ways: (1) when the drug paddles is important to preclude carryover of
dissolution results are not highly variable, and drug or medium.
understanding the effect of an in-residence probe The baskets need special care and examina-
is desired, two concurrent runs (same sampling tion. The basket mesh/screen can become
intervals, n ¼ 6) using manual and automated frayed, misshapen, or warped with use. Screen
sampling methods are compared using the mesh size may change over time, especially
criteria established for intermediate precision, when used with acidic medium. There are
or (2) if the dissolution results are highly variable different designs for attaching baskets to shafts.
(i.e., the RSD is above 20% at time points of The attachment can be with clips or with O-
10 min or earlier and 10% RSD or above at later rings. These attachment variations can affect
time points), the analysis can be performed by dissolution results, depending upon the product;

IV. Specific tests: drug product


18.6 Sources of error in dissolution testing 495
therefore, this factor should be taken into consid- 18.6.3 Vibration and mechanical
eration when evaluating the robustness of a aberration
dissolution method.28 Baskets are especially
prone to gelatin or excipient build-up if not thor- A common problem with dissolution equip-
oughly cleaned immediately after use. ment is vibration,31e33 which can be minimized
The major components of dissolution equip- by ensuring that the top plate and lids for Appa-
ment are the housing for the spindle assembly ratus 1 and 2 are properly leveled. External sour-
(sometimes called the tester head), water bath, ces of vibration include other equipment on
paddles, baskets and shafts, vessels, samplers, bench tops, especially shakers, centrifuges, or
and analyzers. Mechanical aspects of the sonicators. Local construction in the area or
equipment, such as media temperature, paddle within the building is a common, though often
or basket speed, shaft centering and wobble, overlooked, source of vibration. The testers
and vibration can all have a significant impact should not be near hoods or significant air flow
on the rate of dissolution of the drug. sources. Heavy foot traffic and door slamming
also should be avoided. The water bath itself is
rarely a source of vibration because the water
bath designs have evolved to eliminate noisy cir-
18.6.2 Dissolution vessels culators near the bath.
Problems caused by vessel irregularities are Careful maintenance of the equipment is
often overlooked. Vessels are manufactured equally critical. For example, the spindle bear-
from large glass tubing, from which the vessel ings of Apparatus 1 and 2 can become worn
bottom is molded. Depending upon techniques over time, and causing vibration and wobble of
of the molding process, irregular surfaces of the the shaft. In addition, the drive belts should be
vessels can occur and the uniformity of vessel checked regularly for wear and the presence of
bottom roundness can vary. Cheaply made ves- dirt and the tension of belt should be adjusted
sels are notorious for this problem. Close exam- for smooth operation. Although difficult to
ination of vessels when newly purchased is very detect without very close observation of the tes-
important, as surface irregularity can cause ters, surging of spindles can also cause spurious
dissolution results to differ significantly.21,29,30 results. Vessels need to be locked in place so they
Another common problem with vessels is resi- do not move during operation.
due build-up either from oily products or sticky
excipients. Insoluble product, if not rinsed well
from previous testing, can cause contamination
18.6.4 Dissolved gases
in subsequent tests. Vessels that become Dissolved atmospheric gases in the dissolu-
scratched and etched after repeated washing tion medium can adversely affect dissolution,
should be discarded. Lids always need to be and thorough deaeration of the dissolution me-
in place during a dissolution run to prevent dia is essential for generation of reproducible re-
evaporation. As mentioned before, vessels sults. Dissolved oxygen and other gases can
should be locked down to avoid vibration. result in presence of bubbles, which are
The bath water level should always be main- commonly observed in nondeaerated medium.
tained at the top of the vessels to ensure uni- USP General Chapter <711>, Dissolution,5 states
form heating of the medium. Monitoring of that bubbles can interfere with dissolution test
the temperature of the media in all the vessels results and should be avoided. Dissolved air
used in a test (rather than just one) can assure can slow down dissolution by creating a barrier;
the temperature uniformity. either adhering to the tablet surface or to basket

IV. Specific tests: drug product


496 18. Dissolution

screens or particles can cling to bubbles on the areas of testing are especially troublesome. Sam-
glass surface of the vessel or shafts. ple introduction can be difficult and sometimes
The dissolution test should be performed as impossible to control and some products have
soon as practical after deaeration of the medium. dissolution profiles that are “position depen-
It is common practice to rotate the paddle to dent.” For example, if the tablet is off-center,
equilibrate the temperature of the dissolution the dissolution rate may be higher due to shear
medium. However, excessive stirring prior to forces. Conversely, placing the sample close to
starting the experiment can result in redissolu- the center can result in coning and a decrease
tion of atmospheric gasses. The paddle should in dissolution rate. Film-coated tablets can be
always be stopped prior to adding the test sticky when wetted, resulting in inconsistent
article, and great care should be taken to ensure positioning of the test article in the vessel.
that the test article falls in the same place every Replacement of the paddle with a basket or the
time at the bottom of the vessel and does not use of a sinker is the best way to minimize the ef-
stick to the sides. fects of position provided build-up of gelatinous
There are several methods for deaeration of matter does not impair the dissolution.
medium, some manual and some automated. Suspensions can be introduced into the disso-
The method described in USP General lution vessel in a variety of ways: manually, us-
Chapter <711>5 uses heat, filtration, and vac- ing syringes or pipettes, pouring from a tared
uum. Helium sparging is also frequently used beaker, or automated delivery using calibrated
for deaeration of the dissolution medium; how- pipettes. Each method has its own set of limita-
ever, recent shortages of helium have made this tions, although automated sample introduction
approach less practical and commonplace. tends to reduce variability. Mixing of a suspen-
sion sample can generate air bubbles; therefore,
the mixing time of suspension samples must be
18.6.5 Standard solutions strictly controlled to reduce erroneous or biased
Great care should be taken when preparing results.
standard solutions, especially if the standard 18.6.6.2 Dissolution media
must be dried before weighing, as well as
ensuring that the drug powder is completely dis- The dissolution medium is a critical compo-
solved. Notably, prednisone USP RS becomes nent of the test that can cause problems if not
very hard upon drying and difficult to dissolve carefully controlled. One cause of inaccurate re-
in water. Dissolving the powder first in a small sults is the volume of medium withdrawn
amount of alcohol often helps to eliminate this through multiple sampling if not replaced care-
problem. A history of the typical absorptivity fully to maintain sink conditions.
range of the standard can also be very useful to Surfactants can be difficult to clean from the
determine if the standard has been prepared surface of the vessel, especially if the concentra-
properly. tion is high (>0.5%). Surfactants, such as SLS,
can accumulate on the surfaces of the sampling
lines, carboys, other glass containers, and plastic
18.6.6 Method considerations bottles, which may require extensive rinsing to
assure complete removal. Batch-to-batch vari-
18.6.6.1 Sample introduction ability of surfactants, SLS in particular, in terms
The best way to avoid errors and data “sur- of grade, age, impurities (especially electrolytes)
prises” is to put a great deal of effort into the can significantly and affect solubility.34 The
development and validation of methods. Some foaming properties of surfactants can make it

IV. Specific tests: drug product


18.6 Sources of error in dissolution testing 497
very difficult to effectively deaerate the medium automated and manual sampling.37 The filtra-
as well and some peristaltic pumps used in auto- tion step is a critical yet underestimated source
mated equipment are not adequate for surfactant of problems in the dissolution test and needs to
medium. Therefore, deaeration of the dissolution be adequately evaluated with both the standard
medium prior to addition of the surfactant can solution and sample solution. Care should be
reduce some of the problems associated with taken that the filter pore size is not larger than
foaming. One caution when lowering a basket the drug particle size; this is especially important
into surfactant medium is that surface bubbles with micronized or nanoparticles. Filtration also
can adhere to the bottom of the basket and removes insoluble excipients that may otherwise
decrease the dissolution rate substantially. cause a high background.
Several sources of error may be encountered Validation of the prewetting or discard volume
when performing HPLC analysis if surfactants is critical for both the sample and standard solu-
are present in the medium. Auto-injectors may tions. Plugging of filters is a common problem,
need repeated needle washing to be adequately especially with automated sampling devices.
cleaned, and solutions containing high concen- Manual sampling techniques can introduce
trations of surfactants may be too viscous for ac- error by virtue of variations in strength and
curate delivery and may need to be diluted size of the human hand from analyst to analyst.
before injection. Surfactants can also build up Therefore, the pulling velocity through the filter
on the surface of column packing material lead- may vary considerably. Too rapid a movement
ing to extraneous peaks and changes in chro- of liquid through the filter can compromise the
matographic behavior.35 Basic media (pH > 8) filtration process itself.
can cause hydrolysis of some HPLC packing ma-
terials, which can be avoided by either diluting 18.6.6.5 Automation
the samples with a suitable buffer or choosing While automation of dissolution sampling is
a column that can operate at higher pH values. very convenient and laborsaving, errors often
occur with automated devices when potential
18.6.6.3 Sinkers problem areas are overlooked. Sampling lines
Sinkers are defined in USP as “not more than a are often a source of error for a variety of rea-
few turns of a wire helix.” Other sinkers may be sons: unequal lengths between the dissolution
used, but the analyst should be aware of the ef- vessels and the analytical instrument, crimping,
fect different types of sinkers may have on mix- wear beyond limits, disconnection, carryover,
ing.36 Sinkers can be barriers to dissolution line mix-ups or crossing, and inadequate clean-
when the wire is wound too tightly around the ing. The volume dispensed, purged, recycled,
dosage unit. or discarded should be checked routinely.
Pumping tubes can wear out through normal
18.6.6.4 Filters use or repeated organic solvent rinsings and
Filters are used invariably in the preparation may require replacement.
of dissolution samples to prevent undissolved Flow cells in UV-spectrophotometric analysis
particles from entering the analytical sample can be a source of error due to air bubbles that
and further dissolving. Filtration is also essential become caught in the flow cell, either introduced
to prevent the introduction of particulate matter via a water source containing bubbles or by air
into the analytical instrument; this is particularly entering inadvertently into poorly secured sam-
important when the samples are analyzed by ple lines. Flow rate and dwell time should be
HPLC to avoid blockage of the column. Many evaluated, to ensure the absorbance readings
types or different filter materials are used in have reached a steady state. Detector cells need

IV. Specific tests: drug product


498 18. Dissolution

to be cleaned frequently to avoid buildup of dissolution test. Video recording of the dissolu-
drug, excipients, surfactants, or buffer salts tion process can be an invaluable approach in
from the dissolution medium. diagnosing anomalous or poor reproducibility
A common source of error can derive from a of dissolution results. Accurate, meaningful
different filter type being used in the automated dissolution occurs when the product dissolves
dissolution test versus the manual dissolution without disturbance from barriers to dissolution,
test. If the same filter type and pore size cannot or disturbance of vessel hydrodynamics from
be used, then filter validation is essential. any source. The particle disintegration pattern
must show freely dispersed particles. Anoma-
18.6.6.6 Cleaning lous dissolution usually involves one or more
The analyst should take special care to of the following observations:
examine this aspect when validating the method.
• floating chunks of tablet
In many laboratories, where different products
• spinning
are tested on the same equipment, this is a crit-
• coning
ical issue that, if inadequately monitored, may
• mounding
be a cause of inspection failures and erroneous
• gumming
results.
• swelling
18.6.6.7 Method transfer • capping
• “clam shell” erosion
Problems occurring during transfer of
• off-center position
methods can often be traced to not having
• sticking of particles adhering to apparatus or
used exactly the same type of equipment, such
vessel walls
as baskets/shafts, sinkers, dispensing appa-
• sacs
ratus, sampling method, or difference in the
• swollen/rubbery mass
testing environment. The sampling technique
• clear pellicles
(manual vs. automated), and sample introduc-
tion, should be uniform. A precise description Along with good documentation, familiarity
of medium and standard preparation, including with the dissolution behavior of a product is
grade of reagents, in the method is essential and essential in quickly identifying changes in stabil-
should go well beyond simply stating the ity or changes associated with a modification of
method should be conducted according the the formulation. One may notice a change in
relevant pharmacopeial general chapter. Video the size of the dissolving particles, excipients
recording may be useful to detect differences floating upward, or a slower erosion pattern.
between sites. Changes in the formulation or an increase in
strength may produce previously unobserved
basket screen clogging. If the contents of the bas-
18.7 Visual observations ket immediately fall out and settle to the bottom
of the vessel, a spindle assembly surge might be
One of the most useful tools for identifying indicated. If the medium has not been properly
sources of error is close visual observation of deaerated, the analyst may see particles clinging
the dissolution test. Trained analysts can to vessel walls. The presence of bubbles always
pinpoint many problems because they have indicates that deaeration is necessary. Lastly,
developed a knowledge and understanding of the water bath should contain clean water so vi-
the cause and effect relationships of well- sual observations of the dissolution test can be
known issues that can occur during the made clearly and easily.

IV. Specific tests: drug product


18.9 Regulatory aspects and specifications 499

18.8 Performance verification of guidance: “Use of mechanical calibration of dissolu-


dissolution equipment tion apparatus 1 and 2 e current good
manufacturing practice (CGMP).”44
In order for a dissolution method to be consid- This guidance has created considerable con-
ered valid, the dissolution apparatus must be set troversy as it states that an enhanced mechanical
up, qualified, and operated in compliance with calibration (MC) can be used as an alternative
appropriate compendia, as applicable. USP Gen- to the current Apparatus Suitability procedure
eral Chapter <711>, Dissolution, lists apparatus (PVT) for Apparatus 1 and 2 described in the
specifications, the apparatus suitability test USP General Chapter <711>. Both procedures
(now called Performance Verification Testing executed according to a written protocol will
(PVT)), the dissolution medium requirements, satisfy the cGMP requirements for calibration
as well as specific procedure requirements for of laboratory apparatus and mechanical equip-
USP Apparatus 1 and 2.5 The PVT informs the ment for manufacturing, as set forth in CFR xx
analyst whether the equipment is operating 211.160(b) and 211.68, respectively.
properly, and it is always preceded by mechani-
cal calibration. The acceptance criteria of the PVT 18.9 Regulatory aspects and specifications
for Apparatus 1 and Apparatus 2 were revised in
2010. The current acceptance criteria include a The regulatory agencies for the various global
geometric mean and standard deviation. There regions generally address dissolution guidelines
is a “single-stage” test consisting of two consec- in terms of the particular testing necessary to
utive runs of six (or eight, depending on the demonstrate the appropriate or intended release
apparatus’s configuration) and a “two-stage” from a dosage form. These guidelines relate
test in which one run is evaluated and, if it more to the development of an appropriate
does not pass either or both criteria, another dissolution method than the validation proced-
run or runs are performed. A detailed explana- ure itself. As a rule, the specifics of the analytical
tion of these criteria was published in 2009.38 A validation for dissolution procedures are not
“toolkit” that contains a calculation for evalu- separated from the discussions of general
ating whether a PVT passes or fails is available method validation as most of the critical valida-
at the USP website.39 This toolkit also provides tion analysis parameters do not differ between
comprehensive information on how to perform dissolution methods, and, for example, assay
mechanical calibration. methods. As the regional regulatory and ICH
Valid use of the PVT requires choosing either guidelines are discussed in detail elsewhere in
the one-stage or two-stage test before testing be- this book, the reader is referred to those chapters.
gins. The instructions clearly state that choosing Clearly, the final dissolution specification for
which test to use after examining the data invali- a drug product needs to reflect the optimal disso-
dates the use of this compendial tool/worksheet. lution test developed and validated according to
Despite recent efforts of the pharmacopeia to the discussions throughout this chapter. Accept-
standardize procedures to reduce errors, able limits for dissolution results are dependent
including training courses, the major sources of on the type of dosage form and the desired
dissolution variability continue to be vibration, release characteristics, e.g., immediate release
centering, vessel design, and deaeration. A versus extended release.
detailed look at the PVT and industry trends The FDA has provided some guidance on
on the use of mechanical tests and PVT was pub- setting acceptable dissolution specifications
lished in a special edition of Dissolution Technol- that take into account the desired product
ogies in May 2010.40e43 There is also an FDA release characteristics and, in particular, BCS

IV. Specific tests: drug product


500 18. Dissolution

classification considerations.4,26 A recent FDA drug products are rare, the specifications are usu-
guidance shows a further breaking down of im- ally based on dissolution data from pivotal
mediate release formulations to cover specif- batches and other important lots, including stabil-
ically immediate release drug products ity data.
containing high solubility drug substances.18 It
should be noted that the FDA provides only
guidance and the recommendations posed in
the various guidances are not legally binding.
18.9.2 Extended-release drug products
That said, it is recommended that proposed Extended-release formulations are, in essence,
specifications be discussed with FDA prior to designed such that release from the dosage form,
presenting in a formal submission. Specifica- and hence the dissolution of the drug substance,
tions that are within the FDA guidance for a is controlled and occurs over an extended period
specific dosage form are likely to receive less of time. As such, single point, or in most cases
resistance than those which are outside the two-point, specifications are insufficient to
guidance, but in any case, the specifications adequately characterize the quality and batch-
must be justified with appropriate supportive to-batch consistency of the drug product. In
data. All dissolution specifications must addition, because of the designed drug release
apply to a given drug product over its entire characteristics, it is often possible to establish
shelf life. an IVIVC for these formulations. Whether or
not an IVIVC can be established, the specifica-
tion should reflect results from biobatches
designed to determine acceptable clinical
18.9.1 Immediate release drug products
behavior. The dissolution specification for
Specifications for immediate release dosage extended-release formulations should include a
forms are intended to assure a quality measure minimum of three time points, one early time
of batch-to-batch consistency and in some cases point to establish the absence of dose dumping,
an indicator of acceptable bioavailability. For one time point in the middle of the release pro-
drug products containing drug substances with file, and one at the end where at least 80% of
high solubility over the physiological pH range, the dose has been released.
dissolution is primarily used as a quality control Approaches to IVIVC for extended-release
check for batch-to-batch consistency, and a sin- products are discussed in detail in the FDA
gle point specification is most often considered guidance: Extended-release solid oral dosage
sufficient. In such cases, a generally acceptable forms: development, evaluation, application
specification is not less than 85% (Q ¼ 80%) dis- of in vitro/in vivo correlations,45 and USP
solved in 30 min. Acceptance criteria other that General Chapter <1088>, In Vitro and In Vivo
this can be proposed but must be accompanied Evaluation of Dosage Forms46 A well-
by appropriate supportive data.26 established correlation will allow for a reason-
For poorly soluble drugs, a two-point specifi- able prediction of the in vivo behavior of
cation is sometimes needed. It is also possible formulations without performing a bioavail-
that in some cases an IVIVC can be established. ability study. Finding the appropriate correla-
If that is the case, specification limits should be tion has been the focus of numerous
set based on the IVIVC and the dissolution test studies.47e49 The establishment of an IVIVC
can be used to distinguish between bioequivalent has been the subject of much discussion
and bioinequivalent formulations or batches. recently in the arena of QbD and setting clini-
However, since IVIVC for immediate release cally relevant specifications.50

IV. Specific tests: drug product


References 501

18.9.3 Generic drug products 5. Oral Drug Absorption56


6. Poorly Soluble DrugsdDissolution and Drug
There are essentially three scenarios that Release57
guide the approach to setting dissolution specifi- 7. Biopharmaceutic considerations in drug
cations for generic products.26 For many generic product design and in vitro drug product
products, a compendial monograph is available. performance, Applied Biopharmaceutics and
In such cases, it is expected that the generic prod- pharmacokinetics, Chapter 15.20
uct meets the specification provided in the
monograph. If a compendial dissolution test is
not available but a dissolution test for the Refer- References
ence Listed Drug Product (RLD) is publicly
1. USP 18eNF 13; USP: Rockville, MD, USA, 1970.
available, the dissolution test and specification 2. USP 42eNF 37; USP: Rockville, MD, USA, 2018.
should be consistent with the RLD. There is 3. USP 42eNF 37, Validation of Compendial
also an FDA database of dissolution methods Procedures <1225>; USP: Rockville, MD, USA, 2018.
which provides methods FDA prefers for a 4. Waiver of in Vivo Bioavailability and Bioequivalence
drug product.51 If neither a compendial test nor Studies for Immediate-Release Solid Oral Dosage Forms
Based on a Biopharmaceutics Classification System:
a test for the RLD are publicly available, compar- Guidance for Industry; U.S. Department of Health and
ative dissolution testing between the generic Human Services, Food and Drug Administration, Cen-
product and the RLD should be presented and ter for Drug Evaluation and Research (CDER), U.S
specification considerations should include Government Printing Office: Rockville, MD, USA,
testing data under a variety of test conditions 2018.
5. USP 42eNF 37, Dissolution <711> Rockville; USP: MD,
as well as bioequivalence data. USA, 2018.
6. Japanese Pharmacopeia, 17th ed.; Division of Pharmaco-
poeia and Standards for Drugs, Office of Standards
18.10 Conclusions and Guidelines Development, Pharmaceuticals and
Medical Devices Agency (PMDA): Tokyo, Japan, 2016.
7. The British Pharmacopoeia; The Stationary Office: Lon-
This chapter has provided current GMPs in don, UK, 2013.
the area of dissolution testing method validation 8. European Pharmacopoeia, 9th ed.; European Directorate
and method development for the purpose of fil- for the Quality of Medicines and HealthCare, Council
ing drug products with regulatory agencies. of Europe: Strasbourg, France, 2018.
9. Brown, C.; Friedel, H.; Barker, A.; Buhse, L.; Keital, S.;
There are extensive references and further
et al. FIP/AAPS Joint Workshop Report: Dissolution/
reading provided so the reader can research the In Vitro Release Testing of Novel/Special Dosage
subject in more depth if desired. Forms. AAPS PharmSciTech 2011, 12 (2).
10. USP 42eNF 37, Drug Release <724> Rockville; USP: MD,
USA, 2018.
11. Wang, Q.; Fotaki, N.; Mao, Y. Biorelevant Dissolution:
18.11 Bibliography Methodology and Application in Drug Development.
Dissolution Technol. 2009, 16 (3), 6e12.
1. Dissolution Technologies, Questions, and 12. Brown, C. K. Chapter 12, Dissolution Method Develop-
Answers52 ment: an Industry Perspective. In Pharmaceutical Dissolu-
tion Testing; Dressman, J., Kr€amer, J., Eds.; Taylor and
2. Dissolution Theory, Methodology, and
Francis: Boca Raton, FL, 2005.
Testing53 13. Hanson, R.; Gray, V. Chapter 7, Solving Practical Prob-
3. Pharmaceutical Dissolution Testing54 lems, Method Development, and Method Validation. In
4. Handbook of Dissolution Testing, 3rd Handbook of Dissolution Testing, 3rd ed.; Dissolution
Edition55 Technologies, Inc.: Hockessin, DE, 2004.

IV. Specific tests: drug product


502 18. Dissolution

14. Martin, G.; Gray, V. General Considerations for Dissolu- Health and Human Services, Food and Drug Adminis-
tion Methods: Development, Validation, and Transfer. tration, Center for Drug Evaluation and Research
JVT 2011, 17 (1), 8e11. (CDER), U.S Government Printing Office: Rockville,
15. Rohrs, B. R. Dissolution Method Development for MD, USA, 1997.
Poorly Soluble Compounds. Dissolution Technol. 2001, 27. Rohrs, B.; Thamann, T.; Gao, P.; Stelzer, D.; Bergren, M.;
8 (3), 6e12. Chao, R. Tablet Dissolution Affected by a Moisture
16. USP 42eNF 37, the Dissolution Procedure: Development Mediated Solid-State Interaction between Drug and
and Validation <1092> Rockville; USP: MD, USA, 2018. Disintegrant. Pharm. Res. 1999, 16, 1850e1856.
17. Published online: https://www.ich.org/products/ 28. Gray, V.; Beggy, M.; Brockson, R.; Corrigan, N.;
guidelines.html. Verified on 9 September 2019. Mullen, J. A Comparison of Dissolution Results Using
18. Dissolution Testing and Acceptance Criteria for Immediate- O-Ring versus Clipped Basket Shafts. Dissolution Tech-
Release Solid Oral Dosage Form Drug Products Containing nol. 2001, 8 (4), 8e11.
High Solubility Drug Substances; FDA Guidance for In- 29. Tanaka, M.; Fujiwara, H.; Fujiwara, M. Effect of the Irreg-
dustry; U.S. Department of Health and Human Services, ular Inner Shape of a Glass Vessel on Prednisone Dissolu-
Food and Drug Administration, Center for Drug Evalu- tion Results. Dissolution Technol. 2005, 12 (4), 6e14.
ation and Research (CDER), U.S Government Printing 30. Scott, P. Geometric Irregularities Common to the Disso-
Office: Rockville, MD, USA, 2018. lution Vessel. Dissolution Technol. 2005, 12 (1), 18e21.
19. Gray, V. A. Power of the Dissolution Test in Distinguishing 31. Vangani, S.; Flick, T.; Tamayo, G.; Chiu, R.; Cauchon, N.
a Change in Dosage Form Critical Quality Attributes; AAPS Vibration Measurements on Dissolution Systems and
PharmSciTech, 2018. Published online: https://doi.org/ Effects on Dissolution Prednisone Tablets RS. Dissolu-
10.1208/s12249-018-1197-7. tion Technol. 2007, 14 (1), 6e14.
20. Suarez, S.; Marroum, P.; Hughes, M. Chapter 15, Bio- 32. Gao, Z.; Moore, T. W.; Doub, W. H. Vibration Effects on
pharmaceutic Considerations in Drug Product Design Dissolution Tests with USP Apparatus 1 and 2. J. Pharm.
and In Vitro Drug Product Performance. In Applied Bio- Sci. 2008, 97 (8), 3335e3343.
pharmaceutics and Pharmacokinetics; Shargel, L., Yu, A., 33. Collins, C. C. Vibration: What Is it and How Might it Ef-
Eds., 7th ed.; McGraw-Hill Education, 2016. fect Dissolution Testing. Dissolution Technol. 1998, 5 (4),
21. Eaton, J.; Deng, G.; Hauck, W.; Brown, W.; Manning, R.; 16e18.
Wahab, S. Perturbation Study of Dissolution Apparatus 34. Crison, J. R.; Weiner, N. D.; Amidon, G. L. Dissolution
Variables-A Design of Experiment Approach. Dissolu- Media for In Vitro Testing of Water-Soluble Drugs, Effect
tion Technol. 2007, 14 (1), 20e26. of Surfactant Purity and Electrolyte on In Vitro Dissolu-
22. Kikawai, L.; Tran, D.; Hauck, W.; Shah, V.; Stippler, E. tion of Carbamazepine in Aqueous Solutions of Sodium
Effect of Various Operational Parameters on Drug Lauryl Sulfate. J. Pharm. Sci. 1997, 86 (3), 384e388.
Release from a 1 % Hydrocortisone Semisolid Dosage 35. Wuelfing, W.; Kosuda, K.; Templeton, A.; Harman, A.;
Form Using the Vertical Diffusion Cell Apparatus. Mowry, M.; Reed, R. Polysorbate 80 UV/vis Spectral
Dissolution Technol. 2012, 19 (3), 6e12. and Chromatographic CharacteristicsdDefining Bound-
23. Villalobos, R.; Viquez, H.; Hernandez, B.; Ganem, A.; ary Conditions for Use of the Surfactant in Dissolution
Melgoza, L. M.; Young, P. M. Parameters Affection Analysis. J. Pharm. Biomed. Anal. 2006, 41, 774e782.
Drug Release from Inert Matrices. 1: Monte Carlo 36. Soltero, R. A.; Hoover, J. M.; Jones, T. F.; Standish, M.
Simulation. Pharm. Dev. Technol. 2012, 17 (3), 344e352. Effects of Sinker Shapes on Dissolution Profiles.
24. Chowdary, K. P.; Rao, R.; Surya Prakasa, K. A Factorial J. Pharm. Sci. 1989, 78 (1), 35e39.
Study on the Effects of b-cyclodextrin and Poloxamer 37. Lindenberg, M.; Wiegand, C.; Dressman, J. Comparison
407 on the Dissolution Rate of Valsartan from CD Com- of Adsorption of Several Drugs to Typical Filter
plexes and Their Tablets. J. Pharm. Biol.Chem. Sci. 2011, Materials. Dissolution Technol. 2005, 12 (1), 22e25.
2 (3), 438e444. 38. Hauck, W. W.; Manning, R. G.; Cecil, T. L.;
25. Li, H.; Nadig, D.; Kuzmission, A.; Riley, C. Prediction of Brown, W. E.; Williams, R. L. Proposed Change to
the Changes in Drug Dissolution from an Immediate- Acceptance Criteria for Dissolution Performance Verifi-
Release Tablet Containing Two Active Pharmaceutical cation Testing. Pharm. Forum 2007, 33 (3), 574e579.
Ingredients Using an Accelerated Stability Assessment 39. Published online: https://www.usp.org/resources/
Program (ASAPprimeÒ). AAPS Open 2016, 2, 7. compendial-tools. Verified on 16 December 2018.
https://doi.org/10.1186/s41120-016-0010-5. 40. Martin, G. P.; Gray, V. A. Overview of Dissolution In-
26. Guidance for Industry; Dissolution Testing of Immediate- strument Qualification, Including Common Pitfalls.
Release Solid Oral Dosage Forms; U.S. Department of Dissolution Technol. 2011, 18 (2), 6e10.

IV. Specific tests: drug product


References 503
41. Kr€amer, J.; Schwan, R. Practical Aspects of Dissolution Hydrophilic Metoprolol Tartrate. Pharm. Res. 1998,
Instrument Qualification-A European Perspective. 15 (3), 466e473.
Dissolution Technol. 2011, 18 (2), 11e15. 48. Rohrs, B. R. Dissolution Assay Development for In Vitro-
42. Yan, B.; Lu, X.; Lozano, R. Feasibility Study on Qualifi- In Vivo Correlations. Am. Pharm. Rev. 2003, 6 (1), 8e12.
cation of USP Dissolution Apparatus 1 and 2 Using the 49. Retting, H.; Mysicka, J. IVIVC: Methods and Applica-
Enhanced Mechanical Calibration Procedure. Dissolu- tions in Modified-Release Product Development. Disso-
tion Technol. 2011, 18 (2), 17e23. lution Technol. 2008, 15 (1), 6e9.
43. Salt, A.; Glennon, J. Enhanced Mechanical Calibration of 50. Marroum, P. Clinically Relevant Dissolution Methods
Dissolution Test Equipment. Dissolution Technol. 2011, and Specifications. Am. Pharm. Rev. 2012, 15 (1), 36e41.
18 (2), 25e29. 51. Published online: https://www.accessdata.fda.gov/
44. Guidance for Industry; The Use of Mechanical Calibration scripts/cder/dissolution/. Verified on 16 December
of Dissolution Apparatus 1 and 2 e Current Good 2018.
Manufacturing Practice (CGMP); U.S. Department of 52. Marques, M., Brown, W., Eds. Dissolution Technologies
Health and Human Services, Food and Drug Adminis- Questions and Answers; Dissolution Technologies, Inc.:
tration, Center for Drug Evaluation and Research Hockessin, DE, 2010.
(CDER), U.S Government Printing Office: Rockville, 53. Palmieri, A., Ed. Dissolution Theory, Methodology, and
MD, USA, 2010. Testing; Dissolution Technologies, Inc.: Hockessin, DE,
45. Guidance for Industry;; Extended Release Oral Dosage 2007.
Forms: Development, Evaluation, and Application of 54. Dressman, J., Kr€amer, J., Eds. Pharmaceutical Dissolution
in Vitro/in Vivo Correlations; U.S. Department of Testing; Taylor and Francis: Boca Raton, FL, 2005.
Health and Human Services, Food and Drug Adminis- 55. Hanson, R., Gray, V., Eds. Handbook of Dissolution
tration, Center for Drug Evaluation and Research Testing, 3rd ed.; Dissolution Technologies, Inc.: Hockes-
(CDER), U.S Government Printing Office: Rockville, sin, DE, 2004.
MD, USA, 1997. 56. Dressman, J. B., Lennern€as, H., Eds. Oral Drug Absorp-
46. USP 42eNF 37, in Vitro and in Vivo Evaluation of Dosage tion, Prediction and Assessment; Marcel Dekker: AG,
Forms <1088> Rockville; USP: MD, USA, 2018. Basel, Switzerland, 2000.
47. Eddington, N. D.; Marroum, P.; Uppoor, R.; 57. Webster, G., Jackson, J., Bell, R., Eds. Poorly Soluble
Hussain, J.; Augsburger, L. Development and Internal Drugs-Dissolution and Drug Release; Pan Stanford Pub-
Validation of an In Vitro-In Vivo Correlation for a lishing: Singapore, 2017.

IV. Specific tests: drug product

You might also like