Sei sulla pagina 1di 8

Journal of Pharmaceutical Sciences xxx (2018) 1-8

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


journal homepage: www.jpharmsci.org

Commentary

FIP Guidelines for Dissolution Testing of Solid Oral Products


Horst Dieter Friedel 1, *, Cynthia K. Brown 2, Amy R. Barker 2, Lucinda F. Buhse 3,
Susanne Keitel 4, Johannes Kraemer 5, John Michael Morris 6, Christos Reppas 7,
David C. Sperry 2, Kumiko Sakai-Kato 8, Mary P. Stickelmeyer 9, Vinod P. Shah 10
1
Bayer Aktiengesellschaft, Corporate Quality, Berlin, Germany
2
Eli Lilly and Company, Product Research and Development, and Global Quality Laboratories, Indianapolis, Indiana 46285
3
U.S. Food and Drug Administration/CDER/OPQ, White Oak, Maryland 10903
4
EDQM, Strasbourg, France
5
PHAST, Homburg, Germany
6
Former Irish Medicines Board, Dublin, Ireland
7
National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Greece
8
National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
9
Pharmaceutical Consultant, Indianapolis, Indiana
10
Pharmaceutical Consultant, North Potomac, Maryland 20878

a r t i c l e i n f o a b s t r a c t

Article history: Dissolution testing is an important physiochemical test for the development of solid oral dosage forms,
Received 22 June 2018 tablets, and capsules. As a quality control test, the dissolution test is used for assessment of drug product
Revised 3 August 2018 quality and is specified for batch release and regulatory stability studies. In vitro dissolution test results
Accepted 7 August 2018
can often be correlated with the biopharmaceutical behavior of a product.This article provides a sum-
mary of views from major global agencies (Europe, Japan, United States), pharmacopoeias, academia, and
industry. Based on available guidance and literature, this article summarizes highlights for development
Keywords:
automation and validation of a suitable dissolution method, setting appropriate specifications, in vitroein vivo
bioavailability comparison, and how to obtain a biowaiver.
biopharmaceutics classification © 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
system (BCS)
dissolution
drug delivery system(s)
formulation
in vitroein vivo correlation(s) (IVIVC)
solid dosage form(s)
surfactant(s)

Introductory Remarks Dissolution Testing of Solid Oral Products.1,2 The FIP guidelines from
1981 served to provide information to compendial committees
Dissolution testing is an important physiochemical test for the when dissolution methods were being introduced into the
development of solid oral dosage forms, tablets, and capsules. As a compendia. Compendial harmonization projects between Euro-
quality control test, it is used for assessment of drug product quality pean Pharmacopoeia (Ph. Eur.), United States Pharmacopeia (USP),
and is specified for batch release and regulatory stability studies. and Japanese Pharmacopoeia (JP) were underway, when the FIP
In vitro dissolution test results can often be correlated with the guidelines 1995 were published.
biopharmaceutical behavior of a product. Since 1995, regulatory agencies and pharmacopeial committees
The International Pharmaceutical Federation (FIP) is the global have updated dissolution guidances and general chapters to assure
federation representing 4 million pharmacists and pharmaceutical incorporation of current knowledge, information, and re-
scientists worldwide. FIP has previously published Guidelines for quirements. The FIP in vitro dissolution/drug release focus group
revised the recommendations given in the FIP guideline in 1995
considering the modifications to guidelines for bioavailability,
This scientific publication reflects the views of the author(s) and should not be bioequivalence, and in vitro dissolution. Primary modifications
construed to represent views or policies of U.S. Food and Drug Administration and
from the previous guidances include information on harmoniza-
Japan Ministry of Health, Labour and Welfare.
* Correspondence to: Horst Dieter Friedel (Telephone: þ49-3046817922). tion, apparatus information, references for the compendial buffer
E-mail address: horst-dieter.friedel@bayer.com (H.D. Friedel). preparations, and apparatus suitability. The new sections in this

https://doi.org/10.1016/j.xphs.2018.08.007
0022-3549/© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
2 H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8

guidance inform the reader about the process for dissolution apparatus and conditions with an appropriate justification and
method development including information about modified for- demonstration of the discriminatory power of the test.
mulations and discriminating methods. Specification setting and
biowaiver summaries have also been updated. Basket Apparatus
This article summarizes the views from agencies (Europe, Japan,
United States), pharmacopoeias, academia, and industry. Evaluating The basket apparatus can be used for capsules and tablets. The
the information available in literature about dissolution testing and default basket is 40 mesh but other mesh apertures can be used
the experience of the authors, it provides guidance for: with appropriate justification. The basket can be manufactured in
stainless steel, type 316, or other inert material, or it can be gold-
 Development of a suitable dissolution method. plated (Ph. Eur., JP, USP).
 Validation of the dissolution method and determinative step
(analytical test). Paddle Apparatus
 Setting specifications.
 In vitroeIn vivo comparison. The paddle apparatus can be used for capsules and tablets. The
 Biowaiver. paddle can be in 2 pieces (shaft and blades) or it can be constructed
in a single piece. It can be manufactured in stainless steel, type 316,
This article supports pharmaceutical analysts and scientists to or other inert material (Ph. Eur., JP, USP).
develop suitable dissolution test methods to assess product quality
and in vivo performance. Reciprocating Cylinder

Concept of Dissolution Testing This assembly consists of a set of cylindrical, flat-bottomed glass
vessels, a set of glass reciprocating cylinders; inert fittings (stainless
In vitro dissolution testing serves as an important tool for char- steel type 316 or other suitable material) and screens that are made
acterizing the biopharmaceutical quality of a product at different of suitable inert material that are designed to fit the tops and
stages in its life cycle. Dissolution methodology and its application bottoms of the reciprocating cylinder. It can be used with capsules
evolve along with the drug product throughout the development and tablets. As its movement generates more turbulence when
process. In early drug development, in vitro dissolution properties compared to the paddle apparatus, it may be useful for products
are supportive for choosing between alternative formulation can- containing low-solubility drug substances or substances dispersed
didates for further development and for evaluation of active in- in an oily vehicle. The cylinders are arranged in rows and can
gredients (drug substances). In vitro dissolution data are supportive contain dissolution medium with different composition/pH in each
in the evaluation and interpretation of possible risks, especially in row giving the option of running dissolution profiles in different pH
the case of controlled/modified release dosage forms. These include ranges (EP, USP). This apparatus is not described in JP.
dose dumping, food effects on bioavailability, and interaction with
other drugs that might influence gastrointestinal (GI) environ- Flow-Through Cell
mental conditions. Biopharmaceutical aspects are as important
over shelf life as they are for batch release after production; This assembly consists of a reservoir and a pump for medium, a
therefore, in vitro dissolution is of high relevance in quality control flow-through cell, and a water bath that maintains the temperature of
and stability testing. Last but not least, in vitro dissolution data are the medium at the appropriate level during the test. It can accom-
of great importance when assessing changes in production site, modate different sizes and types of cells. There are specific cells for
manufacturing process, or formulation and assist in decisions liquid-filled capsules, granules, powders, etc. The flow rate can be set
concerning the need for conducting supporting bioavailability to work with very low or very high volumes of dissolution medium.
studies. None of these purposes can be fulfilled by an in vitro test Because the apparatus can be configured to allow high volumes of the
system unless it has sufficient reliability. Reliability here means a dissolution medium to reach the dosage form, it can be used with
system that is experimentally sound, yielding precise, accurate, dosage forms containing poorly soluble drug substances. The
repeatable results and with sufficient knowledge of the in vivo apparatus can be operated in an open-loop mode in which the fresh
relevance of the dissolution data obtained. Requirements for medium reaches the sample throughout the entire test, or in a closed-
dissolution testing have been reviewed in the literature. 1-5 Since in vitro loop mode, where the dissolution medium is recirculated.
dissolution is a physical test, defined by convention and is of a The 2 most commonly used dissolution apparatuses are the
destructive nature, proving reliability requires special attention. It basket and paddle apparatuses. The basket and paddle apparatuses
therefore is within the scope of these guidelines to define suitable are simple, robust, and adequately standardized apparatuses that
testing equipment and experimental design, to suggest the background are used universally and thus are supported by the widest experi-
for adequate physical and analytical validation, together with verifi- ence of experimental use. It is because of these advantages that the
cation procedures according to the state of biopharmaceutical science. paddle and rotating basket apparatuses are recommended in
The guidelines are primarily dedicated to solid oral products. However, various guidelines as first choice for the in vitro dissolution testing
the general concepts herein may be adapted to in vitro dissolution of immediate as well as controlled/modified release preparations.
testing of drug substances/powders, semisolid oral products, supposi-
tories and, with certain restrictions, to other nonoral products. Apparatus Suitability

Compendial Dissolution Apparatus Different instrument qualification and suitability methods have
been proposed by the pharmacopeias, regulatory bodies, and
The choice of a suitable dissolution apparatus and conditions standard setting organizations. The FIP focus group recommends
should start with evaluation of the compendial apparatus described establishing the basket and paddle apparatus suitability by
in the Ph. Eur., JP, and USP. If these apparatuses and conditions are not following the mechanical calibration requirements indicated in
discriminative enough, the next step is to evaluate modifications of ASTM E2503-07 (Standard Practice for Qualification of Basket and
the compendial apparatus. The last recourse is to use noncompendial Paddle Dissolution Apparatus) and referenced in U.S. Food and Drug
H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8 3

Administration (FDA) Guidance for Industry: The Use of Mechanical For low-pH media, diluted hydrochloric acid (0.1 M or 0.01 M) is
Calibration of Dissolution Apparatus 1 and 2dCurrent Good used, whereas at higher pH, buffered solutions (acetate and phos-
Manufacturing Practice (January 2010)6 and JP general information phate buffers) are recommended. Although much of the dissolution
(17th edition, March 2016.7 Reference to JP 17th general informa- compendial methods are harmonized,8 buffer preparations are not
tion). If additional system performance information is desired, a included as harmonized properties. It is recommended to conform
performance verification test using USP Reference Standard tablet to the buffer preparations that are consistent with requirements for
or an established in-house reference product can be conducted. Any the country in which the marketing authorization will be held. USP
strict requirement on the use of a specific performance verification buffer solutions are defined in the USP reagents section for buffer
test tablet is not recommended at this time.8 solutions. Ph. Eur. instructions for buffer preparations are defined in
Ph. Eur.5.17.1 “Recommendations on Dissolution Testing” and JP
Harmonization of the Compendial Chapters instruction for buffer preparations in 9.41 “Reagents, Test Solu-
tions.” The influence of the ionic strength on the dissolution
Most of the compendial general chapters on dissolution testing behavior should be investigated during method development.
are harmonized between the Ph. Eur., the JP, and the USP (Ph. Eur., The use of water as the dissolution medium bears the disad-
2017; JP, 2016; USP, 2016). The International Council for Harmoni- vantage that test condition details, such as pH and surface tension,
zation (ICH) recommends that the official pharmacopeial texts, Ph. can vary depending on the source of water and may be changed
Eur. 2.9.3 Dissolution Test for Solid Dosage Forms; JP 6.10 Dissolu- during the dissolution test itself due to the influence of the drug
tion Test; and USP <711 > Dissolution can be used interchangeably products and the (re)absorption of carbon dioxide from air. When
in the ICH regions when using the basket apparatus, the paddle using water as a dissolution medium, purified water is
apparatus, or the flow-through cell. The dissolution test is not recommended.
interchangeable under certain circumstances, for example, when
enzymes are used in the media, for dosage forms referred to as Surfactant
delayed-release, gastro-resistant, or enteric coated nor when ves-
sels greater than 1 L are used. These exemptions are specified in ICH If sink conditions cannot be achieved at any pH in the physio-
Q4B Annex 7.9 logical pH range, surfactants may be added to enhance the solu-
bility of poorly soluble drugs (Biopharmaceutics Classification
Method Development System [BCS] classes 2 and 4). For definition of BCS classes, see
chapter VII Biowaiver.
A dissolution test method for quality control purposes should Surfactants can be either anionic, cationic, zwitterionic, or
meet the following criteria: neutral. Chemically well-defined surfactants, for example, sodium
dodecyl sulfate, should be given preference. Other commonly used
 It must be robust and reproducible over long time periods. surfactants are polysorbates (Tween 20 or 80), polyoxyethylene 23
 It must be easily transferred from laboratory-to-laboratory. lauryl ether (Brij 35) or cetyltrimethyl ammonium bromide.
 It must be able to discriminate between batches with respect to For an overview of commonly used surfactants and information
critical process parameters and critical material attributes, on their critical micelle concentration, see USP chapter <1092>.10
which may have an impact on the in vivo biopharmaceutical (or The concentration of the surfactant should be the minimum
bioavailability) behavior. However, the method should not be amount needed to obtain sink conditions and be justified by solu-
overdiscriminating such that minor differences in the bility data at 37 C to maintain the discriminatory power of the
manufacturing process or incoming material, which do not have dissolution test method. If different dosages of a drug product are
a clinically relevant impact on the in vivo behavior, result in test tested, the same dissolution medium should be used. Thus, the
failure. amount of surfactant added to the dissolution medium should
provide sink condition for the highest dose. The stability of the drug
substance in the chosen dissolution medium must be investigated
Selection of Dissolution Medium at 37 C simulating the dissolution experiment and additionally at
laboratory storage temperature covering the time needed for
Physicochemical characterization of the active pharmaceutical quantitation.
ingredient (API) provides the framework for the dissolution test
and its application for formulation development. Solubility data at Enzymes
37 C in aqueous buffer solutions at different pH values should be
evaluated to be able to select an appropriate dissolution medium Gelatin, in the presence of certain compounds such as aldehydes
providing sink conditions for the API. and/or when exposed to high humidity and temperature, can cross-
Sink conditions are defined as the volume of medium that is at link rendering it insoluble in aqueous solvents. The presence of
least 3 times that required to form a saturated solution of drug cross-linking will alter the in vitro dissolution behavior of gelatin
substance. Sink conditions assure that dissolution is not signifi- capsules and gelatin-coated tablets. The capsule will not open and
cantly limited by solubility characteristics. release its contents into the dissolution medium, or it will open in a
In general, an aqueous medium should be used for the disso- nonuniform way and the dosage form will disintegrate partially or
lution test. It is not recommended to attempt to strictly mimic the not disintegrate at all. Consequently, the dissolution results will fail
physiologic GI environment (e.g., composition of gastric or intes- and/or will have very high variability. This failure may not reflect a
tinal fluid) but to choose the testing conditions as far as is possible failure in vivo.11,12 Where gelatin capsules or gelatin-coated
reasonable, based on the physicochemical characteristics of drug tablets experience dissolution failure due to the presence of cross-
substance, within the range which a drug or dosage form could linking, it is allowed to add proteolytic enzymes to the dissolution
experience after oral administration. Dissolution media typically medium. The use of pepsin for the dissolution medium with pH
selected should be in the physiological pH range of 1-7.5. A higher equal or below 4.0 is recommended, papain or bromelain for the
pH needs to be justified on a case-by-case basis and in general dissolution medium with pH above 4.0 and below 6.8, and
should not exceed pH 8. pancreatin for the dissolution medium with pH equal to or above
4 H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8

6.8. The addition of enzymes into the dissolution medium is not  API properties such as particle size and crystal forms.
accepted by the JP.  Excipient quantity, quality, and grade.
 Manufacturing process parameters such as
Delayed-Release Dosage Forms  Blending time and rate.
 Granulation conditions.
Dissolution testing of delayed-release dosage forms usually  Compression force and dwell time.
comprises an acid stage (in 0.1 M HCl) and a buffer stage (in  Coating conditions (temperature, moisture, and spray rate).
phosphate buffer pH 6.8). For details, see USP <711> or Ph. Eur. The  Stressed samples.
JP uses parallel rather than sequential test methodology.
During product development, discriminatory power is desired to
exhibit differences in the in vitro performance of products and/or
Hydrodynamics
batches after changes in composition or manufacturing. In the market-
supply phase, the goal of dissolution testing is to prove similarity of a
The volume of the dissolution medium should be 500 mL up to
freshly manufactured batch to the model, for which bioavailability
1000 mL for basket and paddle apparatuses with 900 mL being
data have been generated. However, this requires discriminatory po-
historically the most common volume used.
wer of the method with the goal that the dissolution test should be
For dissolution testing of immediate-release (IR) products most
able to differentiate between drug product batches of different quality
commonly apparatus 1 (basket) with a stirring rate of 50-100 rpm
and different in vivo characteristics. On the other hand, a dissolution
or apparatus 2 (paddle) with an agitation speed of 50 or 75 rpm are
method should not be overdiscriminating as explained previously. The
used. In case a formulation exhibits “coning” under the paddle, an
intention of a robust quality control dissolution method is to ensure
increase of the stirring speed from 50 to 75 rpm is typically suffi-
consistent in vivo performance of the product.
cient to reduce coning and leads to less variable results. Other
If discrimination is not observed, then method conditions
agitation rates may be used with justification but should not go
should be revisited to try to achieve discrimination. In some cases,
outside the range 25-150 rpm.
achieving discrimination is not possible. In these cases, adequate
Apparatus 4 (flow-through) can be used as an open system with
experimental work should nevertheless be done to demonstrate
a fresh solution from the reservoir continuously passing through
that discrimination could not be achieved over reasonable ranges of
the cell where the dosage form is initially accommodated. It is also
formulation and process parameters.
possible to operate it as a closed system by recycling a fixed volume
In most cases, for solid oral dosage forms, the compendial
of liquid.13 Two different cells are used for orally administered solid
method development approaches described previously are suffi-
dosage forms, the large cell (22.6 mm internal diameter) and
cient for quality control of the performance of the drug product.
the small cell (12 mm internal diameter). Flow rates up to about
However, in some cases, alternative methods can be justified. In
16 mL/min can be applied; however, physiologically relevant flow
general, compendial methods should be used, unless they are shown
rates are less than 10 mL/min.14 Various types of filters are available
to be insufficient or unnecessary for the intended purpose. One such
(e.g., glass-fiber, cellulose-based). The appropriate filter or combi-
case is outlined in ICH Q6A Decision Tree #7, which states that
nation of filters should be decided with preliminary experiments.
disintegration testing can be suitable for control of rapidly dissolving
(RD), high-solubility drug products, where disintegration is the rate-
Deaeration limiting mechanism.13 Other scenarios may arise on a case-by-case
basis where compendial methods are either unsuitable or imprac-
The significance of deaeration should be investigated by tical. But, in any scenario, the method design principles listed pre-
comparing dissolution data in a deaerated and nondeaerated me- viously must be adhered to: robust, reproducible, transferrable, and
dium. Air bubbles adhering to the dosage unit may alter the able to discriminate. More recently, surrogate dissolution models
dissolution rate especially for poorly soluble drugs. For dissolution based on near-infrared technology have been used to predict in vitro
media containing surfactants, degassing is usually not an option dissolution and can serve as surrogate for traditional dissolution
because of excessive foaming, and the effect of dissolved air on the testing.15 Typically, in these cases, a conventional dissolution
dissolution behavior is minimized by the lower surface tension. method is still used throughout development and for other quality
control purposes such as shelf life stability studies.
Sinkers
Validation Parameters
Sinkers are often used with apparatus 2 where the dosage form
tends to float (e.g., capsules) or sticks to the vessel wall. Several The dissolution procedure has 2 components: the dissolution
sinker types are commercially available. As sinkers can significantly step and the determinative step. Before starting with the validation
influence the dissolution behavior of a dosage form, the type of of the determinative step, the analytical method parameters must
sinker has to be carefully evaluated and described in the dissolution be defined. For the quantification of the dissolved active ingredient,
procedure. Ph. Eur., JP, and USP recommend the use of wire helix normally ultraviolet (UV) detection is used and in some cases, it is
and a small wire basket. The 3 pharmacopoeias also allow the use of necessary to separate the components by high-performance liquid
any validated sinker. chromatography (HPLC)/ultra-HPLC. It is essential to select appro-
priate conditions for the analytical method, where there will not be
Discriminatory Power interferences from other components such as excipients. The vali-
dation of the determinative step is done by following the recom-
The discriminatory power of the dissolution method should be mendations in the ICH guidance on analytical validation Q2.16
investigated simultaneously during formulation and process devel-
opment. The same studies that are used to design the formulation and Specificity
the process can be applied to determine method discrimination.
Factors that should be considered when evaluating the disso- For dissolution testing, the dissolution results must not be
lution procedure include: affected by excipients, other drug substances that may be present in
H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8 5

the product, and possible degradants. Excipients include coatings, during the analytical finish. Selection of the proper filter material is
printing inks, and capsule shells. The specificity evaluation can be important and should be accomplished, and experimentally justi-
done by spiking a placebo version of the finished product with fied, early in the development of the dissolution method. Important
known amounts of the drug substance. The interference should not characteristics to consider when choosing a filter material are type,
exceed 2%. If the placebo interference exceeds 2%, modification or size, and pore size. In addition, use of the correct filter dimensions
replacement of the method may be necessary. Possible method will improve throughput and recovery and reduce clogging. Filter
modifications include, but are not limited to, selecting an alterna- compatibility should be performed as part of method development.
tive wavelength, subtraction of the baseline, correction for the During method validation, the proposed filter types should be
placebo interference, and degradation of the interfering trans- validated for use to demonstrate appropriate recovery (ensure drug
forming absorbance values (e.g., first derivative).12 is not absorbed or adsorbed to the filter).

Linearity and Range Automation

The linearity should be evaluated using at least 5 concentrations When automated equipment is used, a comparison should be
encompassing the entire dissolution profile. In some cases, it may performed between the automated and the manual procedures. This
be helpful to include the upper limit of uniformity of dose. Linearity could be done either by a parallel sampling approach, meaning
is typically calculated by using an appropriate least squares manual sampling, is performed out of the same vessels of the
regression program. In most cases, a square of the correlation co- automated equipment or by an independent sampling approach.
efficient must not be less than 0.98 and the y-intercept must not be Following the independent sampling approach, 2 dissolution runs
importantly different from zero.12 (automated and manual) using the same homogenous sample are
conducted. The comparison of independent dissolution experi-
Accuracy
ments is preferred because the influence of the entire automated
equipmentdnot only the sampling procedure itselfdon the disso-
Accuracy is typically evaluated by preparing multiple samples
lution results is investigated. To be considered comparable, the re-
containing the active ingredient and any other component present
sults from these experiments should not differ by more than 5%
in the dosage form (e.g., excipients, coating materials, capsule shell,
depending on the accuracy and precision of the method, at least for
etc.) in concentration within the linearity range of the method. The
the specified time points, but preferably for all sampling time points.
measured recovery is typically 95%-105% of the amount added.
Special procedures may be necessary when evaluating the accuracy
Justification of Specification
of the acid stage for delayed-release dosage forms.12

Precision Generally for IR (i.e., non modified formulations), a single or 2-


point acceptance criterion is acceptable to control product quality
Precision should be evaluated to include spiking of drug substances where a minimum amount dissolved at the specified time, for
into placebo matrix or evaluation of actual formulations to cover the example, “not less than 80% released in 30 min in the chosen
anticipated concentration range. Repeatability and intermediate pre- dissolution medium, that is, Q ¼ 75%” where Q is the reference
cision should be addressed and reproducibility may be considered. figure quoted in the harmonized compendial test. Of course in
certain cases for highly potent drugs or those with a narrow ther-
Robustness apeutic index, a single-point acceptance criterion may not be
adequate. Further advice on IR products can be obtained in relevant
The robustness of a method is a measure of its capacity to remain regulatory guidance from FDA, European Medicines Agency (EMA),
unaffected by small but deliberate variations of defined parameters and Ministry of Health, Labour and Welfare (Japan) as well as the
and provides an indication of its reliability during normal usage. compendia.
Acceptance criteria should be established to confirm method For extended-release products, the specifications are naturally
robustness with respect to the defined critical method parameters. more complex and demanding.
Robustness studies are performed for the quantitation method Typically, 3-point acceptance criteria will be included in the
(HPLC or UV) and for the dissolution method. specification:
For HPLC quantitation, small deliberate variations, for example,
in pH and composition of the mobile phase, the flow rate, detection 1. An early time point showing that the drug is not rapidly released
wavelength, and column temperature as well as type of stationary from the formulation and therefore should help to provide
phase should be studied. reassurance that rapid release does not occur in vivo after the
For UV quantitation, small deliberate variations, for example, in dose form has been swallowed (“dose dumping”). Such behavior
detection wavelength, baseline correction, pH, and ionic strength of would be contrary to the product design and may present a
the dissolution medium as well as amount of organic solvent in the safety hazard to the patient. A suitable acceptance criterion
reference solutions should be investigated. might take the form of “not more than 20% (±10%) released after
To prove the robustness of the dissolution method, small vari- 1 h.”
ations in pH and surfactant concentration of the dissolution me- 2. A mid-range time point should be specified with an appropriate
dium, temperature, stirring speed, deaeration, sampling technique, acceptance criterion that supports the intended release char-
and tablet placement should be examined. acteristics of the drug from the formulation. For example, this
might be along the lines of “not less than 50% and not more than
Validation of Sample Handling 70% of the nominal dose released after 6 h.” The time here is
chosen to suit the dosage interval proposed and the therapeutic
Filtration objective for the product. Range is typically ±10% or other ranges
can be used if justified by clinical data.
Filtration removes undissolved material, including API that may 3. A final time point is specified showing that drug release from
otherwise interfere with the analytical result or the procedure the formulation is essentially complete and will typically take
6 H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8

the form of “not less than 80% of the drug is released from the
formulation after 8 h.” Again the time point will be chosen and
justified with reference to the intended dosage interval.

Justification of the specifications and in particular the acceptance


criteria will need to be made at the chosen agitation rate and in the
chosen dissolution medium, for simple aqueous dissolution media.
Where surfactants or other substances are added to the dissolution
media, their presence and concentration need to be scientifically
justified based on data generated in the pharmaceutical develop-
ment section of the marketing authorization dossier. In all cases,
justification of specifications should follow relevant regulatory
guidance and most importantly be based on good science.

In VitroeIn Vivo Comparison

An in vitro test for a given drug product serves as the tool for
biopharmaceutical quality, provided that it can distinguish be-
tween “good” and “bad” batches. Good here means “of acceptable
and reproducible in vivo performance” and for oral products “of
acceptable luminal performance.”
With regard to in vitro evaluation of luminal performance of
drug products, that is, to biorelevant in vitro testing,1 a logical hy-
pothesis would be that the closer the in vitro test conditions are to
those in the GI lumen, the better the chances of predicting intra-
luminal dosage form performance. However, depending on the
level and quality of information one is seeking and depending on
certain characteristics of the API, a complete simulation of all as-
pects of luminal conditions may or may not be necessary to eval-
uate drug product performance. In some cases, not only the API
characteristics but also the dosage form characteristics will dictate
the required Level of simulation of luminal conditions.
Figure 1. An overview of the 4 levels of biorelevant media recommended for the
Levels of simulation of luminal conditions have been proposed2
simulation of the luminal environment during development of oral formulations.2
(Fig. 1). Level 0 media are simple aqueous solutions, the pH of
which is adjusted (usually with a buffer) to represent the pH in the
particular location of the GI tract. At this level, the buffer capacity fed state is considered to be the only condition under which the API
may or may not be physiologically relevant, the aim is rather to or the dosage form experiences a highly variable environment
maintain the pH during the course of the experiment. In Level I during its residence in a specific GI region. Therefore, simulation of
media, both the pH and buffer capacity are adjusted to reflect gastric composition in the fed state as a function of time after
physiological values, in fasted and fed state. Bile components are administration is proposed at Level III.2
additionally included in Level II media to reflect the solubilization Composition of media which, depending on the level of simu-
capacity of the luminal fluids, and differences in luminal compo- lation, reflects key luminal characteristics both for the fasted and
sition between the fasted and fed state are addressed. Level III for the fed state has been proposed with dosing conditions similar
media, the most complex compositions, also include proteins, en- to those conditions that are typically applied in bioavailability/
zymes that are usually present in the aqueous phase of luminal bioequivalence studies of oral drug products.2 Such conditions may
contents, and the viscosity effects on drug release. However, pro- differ from those in day-to-day clinical practice. In the latter case,
cesses like adsorption onto particulate matter in the lumen and for example, it may not be worth distinguishing between fasted and
bacterial degradation of dissolved API are not considered in this fed state conditions in the ascending colon, as fed-like conditions
classification. Depending on the location in the GI lumen and are likely to prevail in this region most of the time.
dosing conditions to be simulated, within each Level, the type and
number of media components may be greater or fewer than those
specified in Figure 1. For example, the addition of dietary lipids may Section 1dVerified IVIVC for Regulatory Purposes
be worth considering for Level II simulation of the fed state con-
ditions in the stomach but are not appropriate when simulating Procedures for achieving a verified in vitroein vivo correlation
fasting gastric conditions. Similarly, agents that affect viscosity may (IVIVC) have been described by FDA (for extended-release prod-
be considered for Level III simulation of conditions in the ascending ucts19) and by EMA (for modified-release products20).
colon but have little relevance for Level III simulation of conditions By establishing a meaningful correlation between in vitro
in the fasted upper small intestine. Luminal hydrodynamics are release characteristics and in vivo bioavailability parameters, the
addressed only at the highest Level of simulation (Level III).4 In in vitro dissolution test can serve as a surrogate marker for in vivo
cases where closed in vitro systems are used, it is envisaged that for behavior and thereby confirm consistent therapeutic performance
Level I, Level II, or Level III simulation of conditions in stomach or of batches from routine production.21,22 Although a number of
upper intestine, volumes of about 250 mL and about 500 mL should techniques may be used,21,22 an attempt to develop a level A IVIVC,
be used for fasted state and fed state simulations, respectively.3-5,17 that is, a correlation that represents a point-to-point relationship
In contrast, in the lower intestine, about 200 mL will usually be between in vitro dissolution and the in vivo input rate (e.g., the
appropriate, regardless of dosing conditions.5,18 The stomach in the in vivo dissolution of the drug from the dosage form), is
H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8 7

recommended to be developed and be verified by confirming its 4. Develop IVIVC. Level A, multiple level C or level B IVIVC21,22
predictability.21,22 Both 2-stage approaches (e.g., deconvolution- could be considered. Instead of level A correlation, in vitro
based approaches) and 1-stage approaches (e.g., convolution- data could, alternatively, be coupled with PBPK modeling ap-
based approaches or physiologically based pharmacokinetic proaches for generating the plasma profile.
[PBPK] modeling approaches) are acceptable. Adequacy of pre-
dictability could be evaluated using summary parameters (e.g., the Biowaiver
maximum concentration, area under the curve, etc.). Such sum-
mary parameters calculated from the predicted concentration-time The term biowaiver is applied to a regulatory drug approval
curve are compared with the respective summary parameters for process when the dossier (application) is approved based on evi-
the observed data. The prediction error (PE), defined as %PE ¼ dence of equivalence other than in vivo bioequivalence testing data.
[(observed valueepredicted value)/observed value]  100, is A biowaiver can be applied only for IR solid oral dosage forms with
calculated for each summary parameter. The absolute value of the BCS class 1 and class 3 drug substances. It is not applicable to
PE for all summary parameters should be less than 15% for each narrow therapeutic index drugs. Also, it is not applicable to prod-
formulation and the average PE for all formulations included in the ucts designed to be absorbed in the oral cavity. A biowaiver is
IVIVC developments should be less than 10% for each parameter. generally based on a dissolution test and dissolution profile com-
With a verified IVIVC in place, the number of in vivo studies during parison, for example, similarity factor f2. Biowaivers can be used
product development may be decreased, specifications may be set during development, for line extensions, generic drug product ap-
more easily, and certain regulatory decisions may be facilitated plications, and postapproval changes.
(e.g., scale-up and postapproval changes).21,22 However, a level A BCS is a scientific framework for classifying drug substances
IVIVC cannot serve as a basis for claiming bioequivalence between based on their aqueous solubility and intestinal permeability. BCS
products from different marketing authorization applicants, based classifies the drug substance into 4 classes based on its solubility
on in vitro data only.21 and permeability:

Section 2dIVIVC Comparison for Development Purposes  Class 1: highly soluble/highly permeable (HS/HP).
 Class 2: low solubility/highly permeable (LS/HP).
Immediate-Release Products  Class 3: highly soluble/low permeability (HS/LP).
 Class 4: low solubility/low permeability (LS/LP).
Biopharmaceutical risk should be initially identified. If the risk is
considered to be low, in vitro dissolution data should be collected When combined with the dissolution of the drug product, BCS
using a compendial dissolution apparatus and media simulating key takes into account 3 major factors that govern the rate and extent of
luminal characteristics. If biopharmaceutical risk is considered to be absorption from IR solid oral dosage forms: dissolution, solubility,
high, the impact of formulation and/or of variation in the process of and intestinal permeability.
formulation on in vivo performance should be evaluated. In such The dissolution test should be carried out with 12 units by
case, it is likely that an enabling (bio-enhanced) product is used, and paddle method or by basket method at pH 1.2, 4.5, and 6.8.
evaluation of its luminal performance should not be strictly related Dissolution is described as very RD (VRD) when 85% of the drug
to intraluminal dissolution but more importantly on its ability to dissolves in 15 min or less, RD when 85% of the drug dissolves in
maintain supersaturated levels intraluminally. In vitro evaluation 30 min, and slowly dissolving when it takes more than 30 min for
could be performed using compendial or noncompendial apparatus, 85% of the drug to dissolve. Only those products which are VRD
and testing conditions should reflect the changing of key luminal (BCS class 1 and class 3 drug substances) and RD (BCS class 1 drug
characteristics due to the GI transit.21,22 Provided that disposition substances) are eligible for biowaiver. For biowaiver purpose, the
and elimination characteristics of the API have been previously dissolution profile of the test (multisource) product must always be
defined, in vitro data could be coupled with PBPK modeling ap- compared with the dissolution profile of the reference (compar-
proaches for generating the plasma profile. Alternatively, in vitro ator) product and must meet the similarity criteria of f2 50. There
data may also be directly compared with luminal data in humans. is no need for profile comparison if the product is VRD.
Subsequent to the initial publication of FDA's BCS guidance in
Modified-Release Products 2000,23 WHO,24 EMA,25 Health Canada,26 and later FDA draft BCS
guidance in 201527 have been published, which provides biowaiver
A procedure similar to that recommended by the regulatory for BCS class 1 and class 3 drugs. The guidance documents differ in
agencies19-22 could be applied. Specifically: definitions and requirements, so that harmonization of BCS
guidelines is desirable. In October 2016, the ICH endorsed the
1. Based on plasma profile after administration of an IR product development of a new multidisciplinary guideline ICH M9 to
and pharmacokinetic-pharmacodynamic relationships and on address BCS-based biowaivers.
absorption modeling, define targeted release rates for the
modified-release product. Conclusions
2. Develop about 3 batches of the product with different release
rates within the range of targeted release rates in vivo. Confirm Dissolution testing is the most powerful in vitro test for solid oral
the release rates by performing in vitro dissolution tests. Com- dosage forms to assure product quality and product performance
pendial apparatus are recommended to be used. Exact in vitro including drug bioavailability. With the developments in formula-
testing conditions could vary with the API and type of modified- tion technology and advancing knowledge in dissolution science,
release products under study so that key luminal characteristics there is an increasing challenge in determining appropriate disso-
relating to compositions and times of residence in various lution test methods to meet all objectives: API form and formulation
regions of the GI lumen are simulated. development, quality control, and change control (life cycle man-
3. Perform a clinical study in healthy subjects, for example, single- agement). Advances in instrumentation have resulted in develop-
dose study in healthy adults with the MR products. Inclusion of ment of more robust dissolution apparatus. To support the global
an oral solution or intravenous product will be useful. marketing and availability of drugs, it is essential that regulatory
8 H.D. Friedel et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-8

authorities and pharmacopeias work together to standardize and 14. Fotaki N, Reppas C. The flow through cell methodology in the evaluation
of intraluminal drug release characteristics. Dissolution Technol. 2005;12:
harmonize the dissolution requirements for global applications.
17-21.
15. Pawar P, Wang Y, Keyvan G, Callegari G, Cuitino A, Muzzio F. Enabling real time
References release testing by NIR prediction of dissolution of tablets made by continuous
direct compression (CDC). Int J Pharm. 2016;512:96-107.
16. International Council on Harmonization. Q2(R1)eValidation of analytical pro-
1. Reppas C, Friedel HD, Barker AR, et al. Biorelevant in vitro performance testing
cedures: text and methodology. Available at: www.ich.org. Accessed October
of orally administered dosage forms-workshop report. Pharm Res. 2014;31:
11, 2018.
1867-1876.
17. Koziolek M, Grimm M, Garbacz G, Kühn JP, Weitschies W. Intragastric vol-
2. Markopoulos C, Andreas CJ, Vertzoni M, Dressman J, Reppas C. In-vitro simulation
ume changes after intake of a high-caloric, high-fat standard breakfast in
of luminal conditions for evaluation of performance of oral drug products:
healthy human subjects investigated by MRI. Mol Pharm. 2014;11:1632-
choosing the appropriate test media. Eur J Pharm Biopharm. 2015;93:173-182.
1639.
3. Malagelada JR, Longstreth GF, Summerskill WH, Go VL. Measurement of gastric
18. Diakidou A, Vertzoni M, Goumas K, et al. Characterization of the contents of
functions during digestion of ordinary solid meals in man. Gastroenterology.
ascending colon to which drugs are exposed after oral administration to
1976;70:203-210.
healthy adults. Pharm Res. 2009;26:2141-2151.
4. Klein S, Butler J, Hempenstall JM, Reppas C, Dressman JB. Media to simulate the
19. U.S. Food and Drug Administration, Center for Drug Evaluation and Research.
postprandial stomach I. Matching the physicochemical characteristics of stan-
Guidance for Industry, Extended Release Oral Dosage Forms: Development, Eval-
dard breakfasts. J Pharm Pharmacol. 2004;56:605-610.
€ hlich CP, Giessmann T, et al. Intestinal fluid volumes and transit of uation, and Application of In Vitro/In Vivo Correlations. Rockville, MD: FDA,
5. Schiller C, Fro
CDER; 1997.
dosage forms as assessed by magnetic resonance imaging. Aliment Pharmacol
20. European Medicines Agency. Guideline on Quality of Oral Modified Release
Ther. 2005;22:971-979.
Products EMA/CHMP/QWP/428693/2013. London, UK: EMA; 2014.
6. United States Pharmacopeia. Department of Health and Human Services, U.S.
21. Polster CS, Atassi F, Wu SJ, Sperry DC. Use of artificial stomach-duodenum
Food and Drug Administration, Center for Drug Evaluation and Research.
model for investigation of dosing fluid effect on clinical trial variability. Mol
Guidance for Industry. In: The Use of Mechanical Calibration of Dissolution
Pharm. 2010;7(5):1533-1538.
Apparatus and 2eCGMP. Rockville, MD: FDA, CDER; 2010.
22. Kourentas A, Vertzoni M, Stavrinoudakis N, et al. An in vitro biorelevant
7. The Ministry of Health, Labour and Welfare. JP 17th edition General Information,
gastrointestinal transfer (BioGIT) system for forecasting concentrations in the
Standard Procedure for Mechanical Calibration of Dissolution Apparatus. Tokyo,
fasted upper small intestine: design, implementation, and evaluation. Eur J
Japan: MHLW; 2016.
Pharm Sci. 2016;82:106-114.
8. Brown CK, Buhse L, Friedel HD, et al. FIP position paper summary here. AAPS
23. U.S. Food and Drug Administration, Center for Drug Evaluation and Research.
PharmSciTech. 2009;10(3):924-927.
Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate-
9. International Council on Harmonization. Q4B, evaluation and recommendation
release Solid Oral Dosage Forms Based on a Biopharmaceutics Classification Sys-
of pharmacopoeial texts for use in the ICH regions, Annex7 dissolution test general
tem. Rockville, MD: FDA, CDER; 2000.
chapter. Geneva, Switzerland: ICH; 2010. Available at: www.ich.org. Accessed
24. World Health Organization. Report series 992 WHO expert committee on speci-
October 11, 2018.
fications for pharmaceutical preparations, 49th report, Annex. 7. Geneva,
10. United States Pharmacopeia. USP Chapter <1092>. Rockville, MD: USP; 2016.
Switzerland: WHO; 2015.
11. Marques MRC. Enzymes in the dissolution testing of gelatin capsules. AAPS
PharmSciTech. 2014;15(6):1410-1416. 25. European Medicines Agency. Guideline on the Investigation of Bioequivalence,
12. Gray VA, Cole E, Toma JMD, et al. Use of enzymes in the dissolution testing of CPMP/EWP/QWP/1401/98 Rev. 1. London, UK: EMA; 2010.
gelatin capsules and gelatin-coated tabletserevisions to Dissolution <711> and 26. Health Canada. Biopharmaceutics Classification System Based Biowaiver. Ottawa,
disintegration and dissolution of dietary supplements <2040>. Pharm Forum. Ontario: Health Canada; 2014.
2014;40(6):1-27. Available at: www.usppf.com. Accessed October 11, 2018. 27. U.S. Food and Drug Administration, Center for Drug Evaluation and Research.
13. International Council on Harmonization. Q6Aespecifications for new drug Draft Guidance Waiver of In Vivo Bioavailability and Bioequivalence Studies for
substances and products containing them: chemical substances. Available at: Immediate-Release Solid Oral Dosage Forms Based on a Biopharmaceutics Clas-
www.ich.org. Accessed October 11, 2018. sification System. Rockville, MD: FDA, CDER; 2015.

Potrebbero piacerti anche