Vous êtes sur la page 1sur 18

1088

IN VITRO AND IN VIVO EVALUATION OF DOSAGE FORMS

The Pharmacopeia provides for dissolution and drug release testing in the majority of monographs for solid oral and transdermal dosage forms. In recognition of the sensitivity of dissolution tests, where a valid bioavailability-bioequivalence ( !- "# study is in hand, the policy of this Pharmacopeia has been to give this information dominant consideration in setting dissolution standards. "arly practice was to develop dissolution requirements based on the in vitro performance of clinically successful formulations. $imilarity in dissolution behavior has long been sought from the perspectives of both bioavailability and quality control considerations. It is the goal of the pharmaceutical scientist to find a relationship between an in vitro characteristic of a dosage form and its in vivo performance. The earliest achievable in vitro characteristic thought to portend an acceptable in vivo performance was tablet and capsule disintegration. ! test for disintegration was adopted in %$P &I' (()*+#. !t that time, no quantitative wor, was done in attempting to demonstrate such a relationship, especially in regard to in vivo product performance. -owever, advances in instrumental methods of analysis ultimately opened up prospects for this wor,. The disintegration test was recogni.ed as being insufficiently sensitive by the %$P-/0 1oint Panel on Physiologic !vailability, and in ()23 the Panel directed the identification of candidate articles for the first twelve official dissolution tests that used !pparatus (. The state of science is such that conduct of in vivo testing is necessary in the development and evaluation of dosage forms. !lso, no product, including suspensions and chewable tablets, should be developed without dissolution or drug release characteri.ation where a solid phase e4ists. This chapter sets forth, for products intended for human use, guidelines for characteri.ing a drug that include5 ((# developing in vitro test methods for immediate-release and modified-release dosage forms, (6# designing in vivo protocols, and (7# demonstrating and assessing in vitro-in vivo correlations for modified-release dosage forms. I/ 'IT89 "'!:%!TI9/ ;issolution and ;rug 8elease Testing<=ethod ;evelopment for Immediate-8elease ;osage 0orms ;issolution testing is required for all solid oral Pharmacopeial dosage forms in which absorption of the drug is necessary for the product to e4ert the desired therapeutic effect. "4ceptions are for tablets meeting a requirement for completeness of solution or for rapid ((+ to (* minutes# disintegration for soluble or radiolabeled drugs. The apparatus and procedure conform to the requirements and specifications given in the general chapter ;issolution >(( . ?enerally, e4periments are conducted at 7> . The dissolution medium preferably is deaerated water or, if substantiated by the solubility characteristics of the drug or the formulation, a buffered aqueous solution (typically p- @ to 3# or a dilute acid (+.++( / to +.( / hydrochloric acid# may be used. The usual volume of the medium is *++ to (+++ m:, with the use of greater volumes (up to 6+++ m:# allowed for drugs having limited solubility. The quantity of medium used should be not less than 7 times that required to form a saturated solution of the drug substance. The significance of deaeration of the medium should be determined. !ddition of solutes (i.e., surfactants# and electrolytes to aid in solubili.ation of the drug must be balanced against the loss of the discriminatory power of the test. The use of hydroalcoholic media is generally not favored. The use of such media should be supported by a documented in vitro-in vivo correlation. Aonversely, it should be recogni.ed that this discriminatory power could in some circumstances be e4cessive in that it may result in detection of differences in dissolution that are not clinically significant. The choice of apparatus should be based on ,nowledge of the formulation design and actual dosage form performance in the in vitro test system. $ince dissolution apparatuses tend to become less discriminating when operated at faster speeds, lower stirring speeds should be evaluated and an appropriate speed chosen in accordance with the test data. The most common operating speeds are (++ rpm for !pparatus ( (bas,et# and *+ rpm for !pparatus 6 (paddle# for solid-oral dosage forms and 6* rpm for suspensions. ! @+-mesh screen is used in almost all bas,ets, but other mesh si.es may be used when the need is documented by supporting data. !pparatus 6 is generally preferred for tablets. !pparatus ( is generally preferred for capsules and for dosage forms that tend to float or that disintegrate slowly. ! sin,er, such as a few turns of platinum wire, may be used to prevent a capsule from floating. 9ther types of sin,er devices that achieve minimal coverage of dosage form surface are commercially available. Bhere the use of a sin,er device is employed, it is incumbent on the analyst to assure that the device used does not alter the dissolution characteristics of the dosage form.

;issolution testing should be conducted on equipment that conforms to the requirements in the chapter ;issolution

>((

and

that has been calibrated with both the %$P $alicylic !cid and Prednisone Aalibrator Tablets. The method of analysis should be validated in accordance with the procedures given in the chapter 'alidation of Aompendial Procedures (66* . The test time is generally 7+ to 2+ minutes, with a single time point specification for pharmacopeial purposes. To allow for typical disintegration times, test times of less than 7+ minutes should be based on demonstrated need. Industrial and regulatory concepts of product comparability and performance may require additional time points, and this may also be a feature required for product registration or approval. 0or registration purposes, a plot of percentage of drug dissolved versus time should be determined. "nough time points should be selected to characteri.e adequately the ascending and plateau phases of the dissolution curve. ;issolution test times and specifications usually are established on the basis of an evaluation of dissolution profile data. Typical specifications for the amount of active ingredient dissolved, e4pressed as a percentage of the labeled content (C#, are in the range of >+D to 3+D C dissolved. ! C value in e4cess of 3+D is not generally used, as allowance needs to be made for assay and content uniformity ranges. 0or products containing more than a single active ingredient, dissolution normally should be determined for each active ingredient. Bhere a dissolution test is added to an e4isting monograph, the disintegration test is deleted. -owever, in the case of sublingual preparations, a short disintegration time may be retained as a monograph specification in addition to a dissolution requirement. ;issolution and ;rug 8elease Testing<=ethod ;evelopment for =odified-8elease ;osage 0orms ;rug release testing is required for all modified-release dosage forms in which absorption of the drug is necessary for the product to e4ert the desired therapeutic effect. The apparatus and procedure conform to the requirements and specifications given in the general chapter ;rug 8elease >6@ . The dissolution medium preferably is deaerated water or, if substantiated by the solubility characteristics of the drug or the formulation, buffered aqueous solutions (typically p- @ to 3# or dilute acid (+.++( / to +.( / hydrochloric acid# may be used. ($ee above under ;issolution and ;rug 8elease Testing<=ethod ;evelopment for Immediate-release ;osage 0orms.# 0or modified-release dosage forms, the p-- and surfactant-dependence of the dosage form should be evaluated by in vitro testing in media of various compositions. The volume of medium will vary depending on the apparatus used and the formulation under test. The choice of apparatus should be based on ,nowledge of the formulation design and actual dosage form performance in the in vitro test system. !pparatus ( (bas,et# or !pparatus 6 (paddle# may be more useful at higher rotation frequencies (e.g., the paddle at (++ rpm#. !pparatus 7 (reciprocating cylinder# has been found to be especially useful for bead-type modified-release dosage forms. !pparatus @ (flow cell# may offer advantages for modified-release dosage forms that contain active ingredients having very limited solubility. !pparatus > (reciprocating dis,# has been shown to have application to nondisintegrating oral modified-release dosage forms, as well as to transdermal dosage forms. !pparatus * (paddle over dis,# and !pparatus 2 (cylinder# have been shown to be useful for evaluating and testing transdermal dosage forms. !t least three test times are chosen to characteri.e the in vitro drug release profile for Pharmacopeial purposes. !dditional sampling times may be required for drug approval purposes. !n early time point, usually ( to 6 hours, is chosen to show that potential dose dumping is not probable. !n intermediate time point is chosen to define the in vitro release profile of the dosage form, and a final time point is chosen to show essentially complete release of the drug. Test times and specifications are usually established on the basis of an evaluation of drug release profile data. 0or products containing more than a single active ingredient, drug release should be determined for each active ingredient. Bhere a single set of specifications cannot be established to cover multisource monograph articles, application of a Aase Three standard is appropriate. In Aase Three, multiple drug release tests are included under the same monograph heading, and labeling requirements are included to indicate with which drug release test a specific product complies and, in some cases, the biological performance to be e4pected. ;rug release testing should be conducted on equipment that conforms to the requirements in the chapter ;rug 8elease >6@

and that has been calibrated with the appropriate %$P calibrators. The method of analysis should be validated in accordance with the procedures given in the chapter 'alidation of Aompendial Procedures (66* .

I/ 'I'9 "'!:%!TI9/ 90 =9;I0I";-8":"!$" ;9$!?" 098=$ In evaluating a modified-release product, a fundamental issue is the types of studies that should be performed to give reasonable assurance of safety and efficacy. Bhile providing important information concerning the release characteristics of the drug from the dosage form, at present in vitro studies are most useful for such purposes as monitoring drug product stability and manufacturing process control. The assessment of safety and efficacy of a modified-release dosage form is best achieved through observing in vivo pharmacodynamics or pharmaco,inetics. =oreover, where there is a well-defined, predictive relationship between the plasma concentrations of the drug or active metabolites and the clinical response (therapeutic and adverse#, it may be possible to use plasma drug concentration data alone as a basis for the approval of a modified-release preparation that is designed to replace an immediate-release preparation. The following guidelines are intended to provide guidance in drug substance evaluation and the design, conduct, and evaluation of studies involving modified-release dosage forms. Bhile these guidelines will focus on oral drug delivery systems, the principles may be applicable to other routes of drug administration (e.g., transdermal, subcutaneous, intramuscular, etc.#. Aharacteri.ation of ;rug $ubstance physicochemical properties Physicochemical information necessary to characteri.e the drug substance in a modified-release dosage form should generally be no less than for the drug substance in an immediate-release dosage form. !dditional physicochemical information may be needed on polymorphism, particle si.e distribution, solubility, dissolution rate, stability, and other release-controlling variables of the active drug entity under conditions that may react to the e4tremes of the physiologic environment e4perienced by the dosage form. 0or purposes of this chapter, active drug entity is ta,en to be the official drug substance. pharmaco,inetic properties It is recommended to characteri.e thoroughly the input absorption profile of the active drug entity from a rapidly available preparation (an intravenous solution or oral solution or a well-characteri.ed 0;!-approved immediate-release drug product#, which serves in turn as a reference to evaluate the input profile of the modified-release dosage form. This information together with the biological disposition characteristics for the active drug entity can characteri.e and predict changes in the bioavailability of the drug when input is modified as in the case of the modified-release dosage form. 0or e4ample, if the active drug entity e4hibits saturable first-pass hepatic metabolism, a reduction in systemic availability could result after oral administration if the input rate is decreased. In designing an oral modified-release dosage form, it may be useful to determine the absorption of the active drug entity in various segments of the gastrointestinal tract (particularly in the colon in the case of dosage forms that may release drug in this region#. The effects of food also may be important, and should be investigated. disposition properties The information required to characteri.e the processes of disposition of the active drug entity from a modified-release dosage form should include those generally determined for the same drug in an immediate-release dosage form. This may include the following5 (. ;isposition parameters<clearance, volume of distribution, half-life, mean residence time, or model-dependent or noncompartmental parameters. 6. :inearity or characteri.ation of nonlinearity over the dose or concentration range which could possibly be encountered. 7. !ccumulation. @. =etabolic profile and e4cretory organ dependence, with special attention to the active metabolites and active enantiomers of racemic mi4tures. *. "nterohepatic circulation. 2. Protein binding parameters and dialy.ability. >. The effects of age, gender, race, and relevant disease states. 3. PlasmaEblood ratios.

). ! narrow therapeutic inde4 or a clinical response that varies significantly as a function of the time of day. pharmacodynamic properties Prior to developing a modified-release dosage form, information described below should be gathered. Aoncentration-response relationships should be available over a dose range sufficiently wide to encompass important therapeutic and adverse responses. In addition, the equilibration time 1 characteristics between plasma concentration and effect should be evaluated. These concentration-response relationships should be sufficiently characteri.ed so that a reasonable prediction of the safety margin can be made if dose-dumping from the modified-release dosage form should occur. If there is a well-defined relationship between the plasma concentration of the active drug entity or active metabolites and the clinical response (therapeutic and adverse#, the clinical performance of a new modified-release dosage form could be characteri.ed by plasma concentration-time data. If such data are not available, clinical trials of the modified-release dosage form should be carried out with concurrent pharmaco,inetic-pharmacodynamic measurements. Aharacteri.ation of the ;osage 0orm physicochemical properties The variables employed to characteri.e the physicochemical properties of the active drug entity as it e4ists or is discernible in the dosage form should be the same as those employed to characteri.e the drug substance. $olubility and dissolution profiles over a relevant p- range, usually from p- ( to p- >.@, should be obtained, with particular attention given to the effect of the formulation (as compared to the active drug entity#. Aharacteri.ation of formulations that are insoluble in aqueous systems may require the addition of sodium lauryl sulfate or other surfactant. pharmaco,inetic properties The types of pharmaco,inetic studies that should be conducted are a function of how much is ,nown about the active drug entity, its clinical pharmaco,inetic and biopharmaceutic properties, and whether pharmaco,inetic studies are intended to be the sole basis for product approval. !s a minimum, ((# a single-dose crossover study for each strength of a modified-release dosage form and (6# a multiple-dose, steady-state study using the highest strength of a modified-release dosage form are required to characteri.e the product. $ome appropriate single-dose crossover and multiple-dose steady-state studies are described below. In some modified-release capsule dosage forms, the strengths differ from each other only in the amount of identical beaded material contained in each capsule. In this case, a single-dose and a multiple-dose steady-state study at the highest dosage strength are sufficient. 9ther strengths may be characteri.ed on the basis of comparative in vitro dissolution data. The following pharmaco,inetic studies would be needed for most modified-release dosage forms. $uch studies may, in this instance, constitute the basis for characteri.ation of the dosage form. If approval is to be sought without conducting clinical trials, it is recommended that there be preconsultation with the regulatory authorities to ensure that an adequate database e4ists for such approval. The types of studies generally conducted can be categori.ed as follows. Aase !< Aase ! applies to the original modified-release oral dosage form of an active drug entity already mar,eted in immediate-release form and for which e4tensive pharmacodynamicEpharmaco,inetic data e4ist. $ingle-dose Arossover $tudy< ! single-dose crossover study should include the following treatments5 the modified-release dosage form administered under fasting conditionsF a dosage form that is rapidly available administered under fasting conditionsF and the modified-release dosage form administered at the same time as a high-fat meal (or another type of meal that has potential for causing ma4imum perturbation#. The study of food effects should include provision for control of the fluid inta,e (e.g., 2 to 3 o..# and temperature (e.g., ambient# at the time of drug administration. The dosage form should be administered within * minutes after completion of the meal. If there are no significant differences in the rate or e4tent of bioavailability (!%A, Ama4, and Tma4# as a function of the meal, then additional food effect studies are not necessary. If significant differences in bioavailability are found, it is necessary to define how food affects the modified-release dosage form, as well as how the food-drug effect relates to time.

The purpose of these studies is twofold5 first, to determine whether there is a need for labeling instructions describing special conditions for administration with respect to meals and second, to provide information concerning the pattern of absorption of the modified-release dosage form compared to that of the immediate-release dosage form. The drug input function should be defined for modified-release dosage forms. 2 This will aid in the development of an appropriate in vitro dissolution test. 0or dosage forms that e4hibit high variability, replicate studies are recommended. %se the following guidelines in evaluating food effect. (. If no well-controlled studies have previously defined the effects of a concurrent high-fat meal on an immediate-release dosage form, studies should be performed to determine whether a food effect is a result of problems with the dosage form, i.e., food-related changes in release, or food effects that are unrelated to the dosage form, such as changes in the drugGs absorption from the gastrointestinal tract or changes in the drugGs disposition (i.e., distribution or elimination# that are independent of absorption. The cause of the food effect should be determined by conducting a single-dose crossover study comparing the solution (or immediate-release dosage form# under fed and fasting conditions. If there is no effect of food, then one would conclude that there are problems with the dosage form. If there is an effect of food, then one would conclude that these are unrelated to the dosage form. 6. The effect of timing on the food-drug effect should be tested by performing a four-way crossover study with the modified-release dosage form administered under the following treatment conditions5 fasting, ta,en with a high-fat meal, ( hour before a high-fat meal, and 6 hours after a high-fat meal. 7. If the food effect on an immediate-release dosage form is determined to result from changes in the dissolved drugGs absorption from the gastrointestinal tract or from changes in drug disposition, studies should be designed to define the appropriate relationship between drug dosing and meals. @. !lternative appropriate studies could be conducted if the applicant were to label the drug for administration with a meal that is not fat-loaded. In this case, an alternative meal composition should be considered. *. The entire single-dose, modified-release absorption profile should be monitored. Bhere appropriate (e.g., in a multipledose study# for specific drugs and drug delivery systems, blood samples should be ta,en following brea,fast on the second day, before the second dose is administered. This sampling schedule is particularly important for once-a-day products. 2. 0or delayed-release (enteric-coated# dosage forms, bioavailability studies to characteri.e adequately the food effects and to support the dosing claims stated in the labeling should be performed. =ultiple-dose, $teady-state $tudies< Bhen data demonstrating linear pharmaco,inetics e4ist for an immediate-release dosage form, a steady-state study should be conducted with the modified-release dosage form at one dose rate (preferably at the high end of the usual dose-rate range# using an immediate-release dosage form as a control. !t least three trough-plasma drug concentration (Amin# determinations should be made to ascertain that steady-state conditions have been achieved. Plasma-drug concentration determinations, over at least one dosing interval of the modified-release dosage form, should be made in each phase of the crossover study. It may be preferable (as in the case of rhythmic variation in absorption or disposition of the drug# to measure concentrations over an entire day in each phase. The presence or absence of circadian variation should be verified. The modified-release dosage form should produce an !%A that is equivalent to the immediate-release dosage form. The degree of fluctuation for the modified-release product should be the same as, or less than, that for the immediate-release dosage form given by the approved regimen. !ppropriate concentration measurements should include unchanged drug and major active metabolites. 0or racemic drug entities, consideration should be given to the measurement of the active enantiomers IenantiomerEdiasteriomer distinctionJ. Bhere comparisons of the pharmaco,inetic properties of an immediate-release dosage form at different doses are not available, or where the data show nonlinearity, steady-state crossover studies comparing effects of the modified-release dosage form with those of the immediate-release dosage form should be conducted at two different dose rates5 one at the low end of the recommended dosing range and the second at the high end of the dosing range. In each case, the modified-release dosage form must meet the criteria described in $tudy I with respect to !%A and fluctuations in plasma drug concentrations. If there are significant differences between the modified-release dosage form and the immediate-release dosage form at either the low or the high dosing rate, these data alone are not adequate to characteri.e the product. ;ata can be misleading when obtained from subjects with atypical drug disposition or physiologic characteristics, relative to the target population. Therefore, subject selection should be randomi.ed or from an appropriate target population. If the modifiedrelease dosage form is for use in a specific subpopulation (e.g., for children#, it should be tested in that population. 8egardless of
$T%;H II< $T%;H I<

whether a drug e4hibits linear or nonlinear pharmaco,inetics, the basis for characteri.ation is equivalence of !%A and of the relative degree of fluctuation of concentrations of the modified-release and immediate-release dosage forms. $teady-state studies in selected patient population groups or drug interaction studies may also be necessary, depending upon the therapeutic use of the drug and the types of individuals for whom the modified-release dosage form will be recommended. 0or drugs having narrow therapeutic indices, it may be necessary to perform more e4tensive plasma concentration measurements to determine the potential for unusual drug-release patterns in certain subpopulations. In such studies, it is advisable to perform more than one !%A measurement per patient to assess variability with both the modified-release and the immediate-release dosage forms. Aase < Aase applies to a non-oral, modified-release dosage form of an already mar,eted active drug entity for which e4tensive pharmacodynamicEpharmaco,inetic data e4ist. Aase ! studies (omitting the food effect studies# would be appropriate for the evaluation of a modified-release dosage form designed for a non-oral route of administration if the pattern of biotransformation to active metabolites is identical for the two routes. If the biotransformation patterns are different, then clinical efficacy studies should be performed with the modifiedrelease dosage form. In addition, special studies may be necessary to assess specific ris, factors related to the dosage form (e.g., irritation andEor sensiti.ation at the site of application#. Aase A< Aase A applies to a generic equivalent of an approved modified-release dosage form. ! generic equivalent of an approved modified-release dosage form should be bioequivalent to the standard modified-release dosage form in its rate and e4tent of availability (i.e., !%A, Ama4, Amin, and degree of fluctuation# in crossover single-dose and steady-state studies. 0or an oral modified-release dosage form, the food studies described under Aase ! should also be performed. $tatistical !nalysis !n appropriate statistical method should be selected. ($ee In 'ivo ioequivalence ?uidances (+)+ #.

The currently accepted criteria in the %nited $tates for equivalence for most dosage forms require that the mean pharmaco,inetic parameters of the test dosage form should be within 3+D to (6*D of the reference dosage form using the )+D confidence interval (or, equivalently, the two-sided test procedure, P K +.+*#, and the upper and lower bounds must be within the )+D confidence interval. I/ 'IT89LI/ 'I'9 A988":!TI9/$ The term in vitroLin vivo correlation first appeared in pharmaceutical literature as a result of the awareness of the concepts of bioavailability and of in vitro dissolution rate determinations. The term in vitroLin vivo correlation refers to the establishment of a rational relationship between a biological property, or a parameter derived from a biological property produced by a dosage form, and a physicochemical property or characteristic of the same dosage form. The biological properties most commonly used are one or more pharmaco,inetic parameters, such as Ama4 or !%A, obtained following the administration of the dosage form. The physicochemical property most commonly used is a dosage formGs in vitro dissolution behavior (e.g., percent of drug released under a given set of conditions#. The relationship between the two properties, biological and physicochemical, is then e4pressed quantitatively. Bith the proliferation of modified-release products, it becomes necessary to e4amine the concept of in vitroLin vivo correlation in greater depth. %nli,e immediate-release dosage forms, modified-release products cannot be characteri.ed using a single-time point dissolution test. 0urthermore, with a modified-release product a patient is to e4perience a specific plasma level curve covering a finite time period, usually (6 to 6@ hours. There must be some in vitro means of assuring that each batch of the same product will perform identically in vivo. !n in vitroLin vivo correlation would satisfy this need. Initially it was thought that developing a meaningful correlation for immediate-release dosage forms would be an easier tas, than for modified-release products. -owever, because of the nature of the principles upon which each type is based, it is believed that an in vitroLin vivo correlation is more readily defined for modified-release dosage forms. =odified-8elease ;osage 0orms general considerations

Initial attempts at developing in vitroLin vivo correlations of modified-release products utili.ed the same concepts as those employed with immediate-release dosage forms. Thus, numerous attempts have been made to correlate one or more pharmaco,inetic parameters, determined from in vivo studies of a product, with the amount released in a given time in an in vitro dissolution test. These were essentially single-point correlations. $uch relationships might indicate that increasing or decreasing the in vitro dissolution rate of the modified-release dosage form would result in a corresponding directional change in the productGs performance. -owever, they revealed little about the overall plasma level curve, which is a major factor for drug performance in the patient. The recognition and utili.ation of deconvolution techniques as well as statistical moment calculations represented a major advance over the single-point approach in that these two methodologies utili.e all of the dissolution and plasma level data available to develop the correlations. Therefore, there are at least three correlation techniques (i.e., deconvolution, statistical moment, and single point#, available to the pharmaceutical scientist. There are mar,ed differences in the quality of the correlation obtained with each procedure. Thus, these methods have been categori.ed and are discussed in terms of the advantages of each along with the resulting potential utility as a predictive tool for the pharmaceutical scientist. A988":!TI9/ :"'":$ Three correlation levels have been defined and categori.ed in descending order of usefulness. The concept of correlation level is based upon the ability of the correlation to reflect the entire plasma drug concentrationLtime curve that will result from administration of the given dosage form. It is the relationship of the entire in vitro dissolution curve to the entire plasma level curve that defines the correlation. :evel !< This level is the highest category of correlation. It represents a point-to-point relationship between in vitro dissolution and the in vivo input rate of the drug from the dosage form. This latter factor is sometimes referred to as in vivo dissolution. In such a correlation, the in vitro dissolution and in vivo input rate curves are either directly superimposable or may be made to be superimposable by the use of a constant offset value. The mathematical description for both curves is the same. $uch a procedure is most applicable to modified-release systems that demonstrate an in vitro release rate that is essentially independent of the typical dissolution media usually employed in pharmaceutics. -owever, this is not a requirement for a :evel ! correlation. Bith this correlative procedure, a productGs in vitro dissolution curve is compared to its in vivo input curve (i.e., the curve produced by deconvolution of the plasma level data#. This may be done by use of mass balance model-dependent techniques, such as the BagnerL/elson procedure or the :ooL8iegelman method, or by model-independent, mathematical deconvolution. The advantages of a :evel ! correlation are as follows5
! point-to-point correlation is developed. This is not found with any other correlation level. It is developed using every

plasma level and dissolution point that has been generated. Thus, it reflects the complete plasma level curve. !s a result, in the case of a :evel ! correlation, an in vitro dissolution curve can serve as a surrogate for in vivo performance. Therefore, a change in manufacturing site, method of manufacture, raw material supplies, minor formulation modification, and even product strength using the same formulation can be justified without the need for additional human studies.
! truly meaningful (in vivo indicating# quality control procedure, which is predictive of a dosage formGs performance, is

defined for the dosage form.


The e4tremes of the in vitro quality control standards can be justified by a convolution or deconvolution procedure.

:evel < %tili.es the principles of statistical moment analysis. The mean in vitro dissolution time is compared to either the mean residence time or the mean in vivo dissolution time. :i,e correlation :evel !, :evel utili.es all of the in vitro and in vivo data but is not considered to be a point-to-point correlation because it does not reflect the actual in vivo plasma level curve, since there are a number of different in vivo curves that will produce similar mean residence time values. 0or this reason, unli,e the case of a :evel ! correlation, one cannot rely upon a :evel correlation alone to justify formulation modification, manufacturing site change, e4cipient source change, etc. In addition, in vitro data from such a correlation could not be used to justify the e4tremes of quality control standards. :evel A< This category relates one dissolution time point (t*+D, t)+D, etc.# to one pharmaco,inetic parameter such as !%A, Ama4, or Tma4. It represents a single point correlation. It does not reflect the complete shape of the plasma level, which is the critical factor that defines the performance of modified-release products. $ince this type of correlation is not predictive of actual in vivo product performance, it is generally only useful as a guide in formulation development or as a production quality control procedure. ecause of its obvious limitations, a :evel A correlation has limited usefulness in predicting in vivo drug performance and is subject to the same caveats as a :evel correlation in its ability to support product and site changes as well as justification of quality control standard e4tremes.

developing a correlation This chapter does not define the only procedures for developing an in vitroLin vivo correlation, as any well-designed and scientifically valid approach would be acceptable. To assist the pharmaceutical scientist, one possible procedure for developing a :evel ! correlation is described below.
The plasma level or urinary e4cretion data obtained in the definitive bioavailability study of the modified-release dosage

form are treated by a deconvolution procedure. The resulting data may represent the drug input rate of the dosage form. It is also considered to represent in vivo dissolution when the rate-controlling step of the dosage form is its dissolution rate (i.e., drug absorption, after it has dissolved, is considered to be instantaneous#. !ny deconvolution procedure (i.e., mass balance or mathematical deconvolution# will produce acceptable results.
The biobatch3 is subjected to in vitro dissolution evaluation, and the effect of varying the dissolution conditions

investigated. $ome of the variables that can be studied are the apparatus (it is preferable to use official apparatus#, mi4ing intensity, and dissolution medium (p-, en.ymes, surfactants, osmotic pressure, ionic strength, etc.#. It is not always necessary to study the dosage formGs dissolution behavior under all of the conditions indicated. The number of conditions investigated will depend largely on whether a correlation can be found with the in vitro results obtained under the more commonly investigated conditions such as apparatus, agitation intensity, or dissolution medium and p- value. "ach formulation and every drug represents an individual challenge. The in vitro evaluation of the dosage form should be performed regardless of the correlation level being developed.
The in vitro dissolution curve is then compared to the drug input rate curve. This can be performed by various methods.

$imply positioning one curve on the other can often indicate the e4istence of a correlation. This may then be quantified by defining the equation for each curve and comparing the corresponding constants. The simplest way to demonstrate a correlation is to plot the fraction absorbed in vivo versus the fraction released in vitro. Bith a :evel ! correlation, this relationship is often linear with a slope of (. The intercept may or may not be + depending upon whether there is a lag time before the system begins to release drug in vivo, or the absorption rate is not instantaneous resulting in the presence of some finite quantity of dissolved but unabsorbed drug. In either case, it is a point-to-point or a :evel ! correlation when the relationship is linear with a slope of (. This indicates that the curves are essentially superimposable.
If from the studies indicated in the in vitro dissolution evaluation above, the modified-release dosage form e4hibits

dissolution behavior that is independent of the variables studied, and a :evel ! correlation is demonstrated when the in vitro dissolution curve is compared to the drug input rate curve, it is li,ely that the correlation is general and can be e4trapolated within a reasonable range for that formulation of the active drug entity. If, however, the dosage form e4hibits dissolution behavior that varies with the in vitro conditions, it must be determined which set of dissolution conditions best correlates with in vivo performance. 9ne can then establish whether the correlation is real or an artifact. This is achieved by preparing at least two formulations having significantly different in vitro behavior. 9ne should demonstrate a more rapid release and the other a slower release than the biobatch. ! pilot !- " study should be performed with these formulations, and the previously established correlation demonstrated for both. The formulation modifications of these batches should be based upon formulation factors that would be e4pected to influence the productGs modified-release mechanism, and modification of these formulation factors are e4pected to influence the dosage formGs release rate.
9nce a :evel ! correlation is established, it is possible that in vitro testing may be utili.ed for establishing the effects of

manufacturing modifications such as minor formulation changes, manufacturing site and equipment change, alternative e4cipient suppliers, and a change in dosage form strength in the same formulation. It is questionable whether such an e4trapolation with :evel and A correlations is possible. "stablishment of ;issolution $pecification 8anges It is relatively easy to establish a multipoint dissolution specification for a modified-release dosage form. The dissolution behavior of the biobatch may be used to define the amount to be released at each time point. The difficulty arises in the variation to be allowed around each time point. In the case of a :evel ! correlation, this may be done in two ways, both of which utili.e the in vitro-in vivo correlation5 convolution and deconvolution. convolution 8easonable upper and lower dissolution values are selected for each time point established from the biobatch. -istorically, dissolution specifications have been selected by using the average dissolution of the development batches, with a range of M 6.* to 7 standard deviations. It is now e4pected that the average dissolution values are appro4imately the same as those of the biobatch. The dissolution curves defined by the upper and lower e4tremes are convoluted to project the anticipated plasma level curves that would result from administration of these formulations to the same panel to which the biobatch was administered. If the resulting plasma level data fall within the )*D confidence intervals obtained in the definitive !- " study, these ranges can

be considered to be acceptable. !n alternative acceptance approach that has been suggested is that when the therapeutic window for a drug has been defined, one may establish an upper and lower limit if the convolution results fall within the therapeutic window, even if they fall outside the confidence interval. If they fall outside the intervals, a more limited range must be established. This should be continued until the predicted values meet the desired ranges. deconvolution !n acceptable set of plasma-level data is established both for a batch of material demonstrating a more rapid release and for one demonstrating a slower release than that of the biobatch. These may be selected by using the e4tremes of the )*D confidence intervals or M( standard deviation of the mean plasma level. These curves are then deconvoluted, and the resulting input rate curve is used to establish the upper and lower dissolution specifications at each time point. In the case of :evel and A correlations, batches of product must be made at the proposed upper and lower limits of the dissolution range, and it must be demonstrated that these batches are acceptable by performing a !- " study. Immediate-8elease ;osage 0orms general considerations $ince the mechanisms for release of drug from modified-release dosage forms are more comple4 and variable than those associated with immediate-release dosage forms, it would be anticipated that an in vitro-in vivo correlation would be easier to develop with the later formulations. %nfortunately, most of the correlation efforts to date with immediate-release dosage forms have been based on the correlation :evel A approach, although there also have been efforts employing statistical moment theory (:evel #. !lthough it is conceivable that the same :evel ! correlation approach may be utili.ed with immediate-release dosage forms, until data have been gathered to support this concept, :evel and :evel A are the best approaches that can be recommended with these dosage forms. ( "quilibration time is a measure of the time-dependent discontinuity between measured plasma concentrations and measured effects. The discontinuity is more often characteri.ed by the degree of hysteresis observed when the effect-concentration plot for increasing concentrations is compared with that for decreasing concentrations. Bhere the equilibration time is very short (i.e., rapid equilibration with no active metabolites generated#, there will be little or no hysteresis. That is, the same effect will be observed for a given concentration independent of the interval between the time of dosing and the time that measurements are made. 6 BagnerL/elson, :ooL8iegelman, and other deconvolution methods are found in te4tboo,s on biopharmaceutics. 7 The batch that was used in the pivotal bioavailability study. !u4iliary Information< Please chec, for your question in the 0!Cs before contacting %$P. TopicECuestion ?eneral Ahapter Aontact Billiam ". rown $enior $cientist (-7+(-3(2-373+ "4pert Aommittee ( PA+*# iopharmaceutics+*

%$P76L/06> Page **6 (+)+ I/ 'I'9 I9"C%I'!:"/A" ?%I;!/A"$ 1

This chapter is divided into two sections that provide guidances related to the general conduct of bioequivalence tests and to bioavailability protocols for specific drugs. These statements were prepared by the 0ood and ;rug !dministration, 9ffice of ?eneric ;rugs (0;!, 9?;#, ;ivision of ioequivalence, and presented herein. 0or general bac,ground information on related issues, refer to In 'itro and In 'ivo "valuation of ;osage 0orms (+33 .

1092

THE DISSOLUTION PROCEDURE: DEVELOPMENT AND VALIDATION

The %$P dissolution procedure is a performance test applicable to many dosage forms. It is one test in a series of tests that constitute the dosage formGs public specification (tests, procedures for the tests, acceptance criteria#. To satisfy the performance test, %$P provides the general test chapters ;isintegration >+( , ;issolution >(( , and ;rug 8elease >6@ . These chapters provide information about conditions of the procedure. 0or dissolution, these include information about ((# medium, (6# apparatusEagitation rate, (7# study design, (@# assay, and (*# acceptance criteria. 9verall the dissolution procedure yields data to allow an acceptEreject decision relative to the acceptance criteria, which are frequently based on a regulatory decision. This chapter provides recommendations on how to develop and validate a dissolution procedure. ?"/"8!: A9=="/T$ The dissolution procedure requires an apparatus, a dissolution medium, and test conditions that provide a method that is discriminating yet sufficiently rugged and reproducible for day-to-day operation and capable of being transferred between laboratories. The acceptance criteria should be representative of multiple batches with the same nominal composition and manufacturing process, typically including ,ey batches used in pivotal studies, and representative of performance in stability studies. The procedure should be appropriately discriminating, capable of distinguishing significant changes in a composition or manufacturing process that might be e4pected to affect in vivo performance. It is also possible for the procedure to show differences between batches when no significant difference is observed in vivo. This situation requires careful evaluation of whether the procedure is too sensitive or appropriately discriminating. !ssessing the results from multiple batches that represent typical variability in composition and manufacturing parameters may assist in this evaluation. It is sometimes valuable to intentionally vary manufacturing parameters, such as lubrication, blend time, compression force, or drying parameters, to further characteri.e the discriminatory power of the procedure. Bith regard to stability, the dissolution test should appropriately reflect relevant changes in the drug product over time that are caused by temperature, humidity, photosensitivity, and other stresses. ! properly designed test should result in data that are not highly variable and should not be associated with significant analytical solution stability problems. -igh variability in results can ma,e it difficult to identify trends or effects of formulation changes. ;issolution results may be considered highly variable if the relative standard deviation (8$;# is greater than 6+D at time points of (+ minutes or less and greater than (+D 8$; at later time points. 1 -owever, most dissolution results e4hibit less variability than this. The source of the variability should be investigated when practical, and attempts should be made to reduce variability whenever possible. The two most li,ely causes are the formulation itself (e.g., drug substance, e4cipients, or manufacturing process# or artifacts associated with the test procedure (e.g., coning, tablets stic,ing to the vessel wall or bas,et screen#. 'isual observations are often helpful for understanding the source of the variability and whether the dissolution test itself is contributing to the variability. !ny time the dosage contents do not disperse freely throughout the vessel in a uniform fashion, aberrant results can occur. ;epending on the problem, the usual remedies include changing the apparatus type, speed of agitation, or deaerationF consideration andEor e4amination of sin,er typeF and changing the composition of the medium. =odifications to the apparatus may also be useful, with proper justification and validation. =any causes of variability can be found in the formulation and manufacturing process. 0or e4ample, poor content uniformity, process inconsistencies, a reaction ta,ing place at different rates during dissolution, e4cipient interactions or interference, film coating, capsule shell aging, and hardening or softening of the dosage form on stability may be sources of variability and interferences. ;uring routine testing of the product, variability outside the e4pected range should be investigated from analytical, formulation, and processing perspectives. =";I%= Physical and chemical data for the drug substance and dosage unit need to be determined before selecting the dissolution medium. Two ,ey properties of the drug are the solubility and solution state stability of the drug as a function of the p- value. Bhen selecting the composition of the medium, the influence of buffers, p- value, and surfactants on the solubility and stability of the drug need to be evaluated. Ney properties of the dosage unit that may affect dissolution include release mechanism

(immediate, delayed, or modified# and disintegration rate as affected by hardness, friability, presence of solubility enhancers, and presence of other e4cipients. ?enerally, when developing a dissolution procedure, one goal is to have sin, conditions, defined as the volume of medium at least three times that required in order to form a saturated solution of drug substance. Bhen sin, conditions are present, it is more li,ely that dissolution results will reflect the properties of the dosage form. ! medium that fails to provide sin, conditions may be acceptable if it is shown to be more discriminating or otherwise appropriately justified. %sing an aqueousLorganic solvent mi4ture as a dissolution medium is discouragedF however, with proper justification this type of medium may be acceptable. Purified water is often used as the dissolution medium, but is not ideal for several reasons. 0irst, the quality of the water can vary depending on the source of the water, and the p- value of the water is not controlled. $econd, the p- value can vary from day to day and can also change during the run, depending on the active substance and e4cipients. ;espite these limitations, water is ine4pensive, readily available, easily disposed of, ecologically acceptable, and suitable for products with a release rate independent of the p- value of the medium. The dissolution characteristics of an oral formulation should be evaluated in the physiologic p- range of (.6 to 2.3 ((.6 to >.* for modified-release formulations#. ;uring method development, it may be useful to measure the p- before and after a run to discover whether the p- changes during the test. $election of the most appropriate conditions for routine testing is then based on discriminatory capability, ruggedness, stability of the analyte in the test medium, and relevance to in vivo performance, where possible. Typical media for dissolution may include the following (not listed in order of preference#5 dilute hydrochloric acid, buffers in the physiologic p- range of (.6 to >.*, simulated gastric or intestinal fluid (with or without en.ymes#, water, and surfactants (with or without acids or buffers# such as polysorbate 3+, sodium lauryl sulfate, and bile salts. The molarity of the buffers and acids used can influence the solubili.ing effect, and this factor may be evaluated. 0or compounds with high solubility and high permeability (as defined by the iopharmaceutics Alassification $ystem#, the choice of medium and apparatus may be influenced by the referenced 0;! ?uidance 1. 0or very poorly soluble compounds, aqueous solutions may contain a percentage of a surfactant (e.g., sodium lauryl sulfate, polysorbate, or lauryldimethylamine o4ide# that is used to enhance drug solubility. The need for surfactants and the concentrations used can be justified by showing profiles at several different concentrations. $urfactants can be used either as wetting agents or to solubili.e the drug substance. 'olume /ormally, for bas,et and paddle apparatus, the volume of the dissolution medium is *++ m: to (+++ m:, with )++ m: as the most common volume. The volume can be raised to between 6 and @ :, using larger vessels and depending on the concentration and sin, conditions of the drugF justification for this procedure is e4pected. ;eaeration The significance of deaeration of the medium should be determined, because air bubbles can interfere with the test results, acting as a barrier to dissolution if present on the dosage unit or bas,et mesh. 0urther, bubbles can cause particles to cling to the apparatus and vessel walls. 9n the other hand, bubbles on the dosage unit may increase buoyancy, leading to an increase in the dissolution rate, or may decrease the available surface area, leading to a decrease in the dissolution rate. ! dearation method is described as a footnote in the Procedure section under ;issolution >(( . Typical steps include heating the medium, filtering, and drawing a vacuum for a short period of time. 9ther methods of deaeration are available and in routine use throughout the industry. =edia containing surfactants are not usually deaerated because the process results in e4cessive foaming. To determine whether deaeration of the medium is necessary, results from dissolution samples run in nondeaerated medium and deaerated medium should be compared. "n.ymes The use of en.ymes in the dissolution medium is permitted in accordance with ;issolution occur as a result of cross-lin,ing with gelatin capsules or gelatin-coated products. In 'itroLIn 'ivo Aorrelation (I'I'A# >(( when dissolution failures

!n in-depth discussion on I'I'A can be found in In 'itro and In 'ivo "valuation of ;osage 0orms discussion follows.

(+33 . ! brief

iorelevant medium is a medium that has some relevance to the in vivo performance of the dosage unit. Ahoice of a biorelevant medium is based on ((# a mechanistic approach that considers the absorption site, if ,nown, and (6# whether the rate-limiting step to absorption is the dissolution or permeability of the compound. In some cases, the biorelevant medium will be different from the test conditions chosen for the regulatory test, and the time points are also li,ely to be different. If the compound dissolves quic,ly in the stomach and is highly permeable, gastric emptying time may be the rate-limiting step to absorption. In this case, the dissolution test should demonstrate that the drug is released quic,ly under typical gastric (acidic# conditions. 9n the other hand, if dissolution occurs primarily in the intestinal tract (e.g., for a poorly soluble, wea, acid#, a higher p- range (e.g., simulated intestinal fluid with a p- of 2.3# may be more appropriate. The fed and fasted states may also have significant effects on the absorption or solubility of a compound. Aompositions of media that simulate the fed and fasted states can be found in the literature. These media reflect changes in p-, bile concentrations, and osmolarity after meal inta,e and therefore have a composition different from that of typical compendial media. They are primarily used to establish in vitroLin vivo correlations during formulation development and to assess potential food effects and are not intended for quality control purposes. 0or quality control purposes, the substitution of natural surfactants (bile components# with appropriate synthetic surfactants is permitted and encouraged because of the e4pense of the natural substances and the labor-intensive preparation of the biorelevant media. !PP!8!T%$E!?IT!TI9/ !pparatus The choice of apparatus is based on ,nowledge of the formulation design and the practical aspects of dosage form performance in the in vitro test system. 0or solid oral dosage forms, !pparatus ( and !pparatus 6 are used most frequently. Bhen !pparatus ( or 6 is not appropriate, another official apparatus may be used. !pparatus 7 (8eciprocating Aylinder# has been found to be especially useful for bead-type modified-release dosage forms. !pparatus @ (0low-Through Aell# may offer advantages for modified-release dosage forms that contain active ingredients with limited solubility. In addition, !pparatus 7 or !pparatus @ may have utility for soft gelatin capsules, bead products, suppositories, or poorly soluble drugs. !pparatus * (Paddle over ;is,# and !pparatus 2 (8otating Aylinder# have been shown to be useful for evaluating and testing transdermal dosage forms. !pparatus > (8eciprocating -older# has been shown to have application to nondisintegrating oral modifiedrelease dosage forms, as well as to transdermal dosage forms. $ome changes can be made to the apparatusF for e4ample, a bas,et mesh si.e other than the typical @+-mesh bas,et (e.g., (+, 6+, 3+ mesh# may be used when the need is clearly documented by supporting data. In countries where available mesh si.es vary from the %$P-specified mesh value, bas,et material with the nearest metric dimension should be used. Aare must be ta,en that bas,ets are uniform and meet the dimensional requirements specified under ;issolution >(( . If the bas,et screens become clogged during dissolution of capsule or tablet formulations, it may be advisable to switch to the paddle method. The volume can be increased from the typical )++ to (+++ m: by using 6- and @-: vessels to assist in meeting sin, conditions for poorly soluble drugs. ! noncompendial apparatus may have some utility with proper justification, qualification, and documentation of superiority over the standard equipment. 0or e4ample, a small-volume apparatus with mini paddles and bas,ets may be considered for lowdosage strength products. The rotating bottle or static tubes (jac,eted stationary tubes enclosed with a water jac,et and equipped with a magnetic stirrer# may also have utility for microspheres and implants, pea, vessels for eliminating coning, and modified flow-through cells for special dosage forms, including powders and stents. $in,ers Bhen sin,ers are used, a detailed description of the sin,er must be stated in the written procedure. It may be useful to evaluate different sin,ers, recogni.ing that sin,ers can significantly influence the dissolution profile of a dosage unit. Bhen transferring the procedure, the sin,ers should be duplicated as closely as possible in the ne4t facility. There are several types of commercially available sin,ers. ! method for ma,ing sin,ers by hand, sin,ers that are similar to Oa few turns of wire heli4P as described in !pparatus 6 (Paddle !pparatus# under ;issolution >(( , is described below. =aterials< %se 7(2 stainless steel wire or other inert material, typically +.+76 inchE6+ gaugeF and cylinders of appropriate diameter (e.g., cor, borers#. $i.es are shown in the accompanying table.

Aapsule :ength of ;iameter Aor, ore $hell Type Bire (cm# $i.e (cm# /umber Q+, elongated Q( and Q6 Q7 and Q@ (6 (+ 3 +.3 +.> +.** @ 7 6

Procedure< Aut the specified length of wire, coil around a cylinder of the appropriate si.e, and use small pliers to curve in the ends. %se caution, because wire ends may be rough and may need to be filed. If the sin,er is handmade, the sin,er material and construction procedure instructions should be documentedF if a commercial sin,er is used, the vendor part number should be reported. !gitation 0or immediate-release capsule or tablet formulations, !pparatus ( (bas,ets# at (++ rpm or !pparatus 6 (paddles# at *+ or >* rpm are most commonly used. 9ther agitation speeds and apparatus are acceptable with appropriate justification. 8ates outside 6* to (*+ rpm are usually inappropriate because of the inconsistency of hydrodynamics below 6* rpm and because of turbulence above (*+ rpm. !gitation rates between 6* and *+ rpm are generally acceptable for suspensions. 0or dosage forms that e4hibit coning (mounding# under the paddle at *+ rpm, the coning can be reduced by increasing the paddle speed to >* rpm, thus reducing the artifact and improving the data. If justified, (++ rpm may be used, especially for e4tended-release products. ;ecreasing or increasing the apparatus rotation speed may be justified if the profiles better reflect in vivo performance andEor the method results in better discrimination without adversely affecting method reproducibility. $election of the agitation and other study design elements for modified-release dosage forms is similar to that for immediaterelease products. These elements should conform to the requirements and specifications given in ;issolution >(( when the apparatus has been appropriately calibrated. $T%;H ;"$I?/ Time Points 0or immediate-release dosage forms, the duration of the procedure is typically 7+ to 2+ minutesF in most cases, a single time point specification is adequate for Pharmacopeial purposes. Industrial and regulatory concepts of product comparability and performance may require additional time points, which may also be required for product registration or approval. ! sufficient number of time points should be selected to adequately characteri.e the ascending and plateau phases of the dissolution curve. !ccording to the iopharmaceutics Alassification $ystem referred to in several 0;! ?uidances, highly soluble, highly permeable drugs formulated with rapidly dissolving products need not be subjected to a profile comparison if they can be shown to release 3*D or more of the active drug substance within (* minutes. 0or these types of products a one-point test will suffice. -owever, most products do not fall into this category. ;issolution profiles of immediate-release products typically show a gradual increase reaching 3*D to (++D at about 7+ to @* minutes. Thus, dissolution time points in the range of (*, 6+, 7+, @*, and 2+ minutes are usual for most immediate-release products. 0or rapidly dissolving products, including suspensions, useful information may be obtained from earlier points, e.g., * to (+ minutes. 0or slower-dissolving products, time points later than 2+ minutes may be useful. ;issolution test times for compendial tests are usually established on the basis of an evaluation of the dissolution profile data. $o-called infinity points can be useful during development studies. To obtain an infinity point, the paddle or bas,et speed is increased at the end of the run for a sustained period (typically (* to 2+ minutes#, after which time an additional sample is ta,en. !lthough there is no requirement for (++D dissolution in the profile, the infinity point can provide data that may supplement content uniformity data and may provide useful information about formulation characteristics during initial development or about method bias.

0or an e4tended-release dosage form, at least three test time points are chosen to characteri.e the in vitro drug release profile for Pharmacopeial purposes. !dditional sampling times may be required for drug approval purposes. !n early time point, usually ( to 6 hours, is chosen to show that there is little probability of dose dumping. !n intermediate time point is chosen to define the in vitro release profile of the dosage form, and a final time point is chosen to show the essentially complete release of the drug. Test times and specifications are usually established on the basis of an evaluation of drug release profile data. 0or products containing more than a single active ingredient, drug release is to be determined for each active ingredient. 9bservations 'isual observations and recordings of product dissolution and disintegration behavior are very useful because dissolution and disintegration patterns can be indicative of variables in the formulation or manufacturing process. To accomplish visual observation, proper lighting (with appropriate consideration of photodegradation# of the vessel contents and clear visibility in the bath are essential. ;ocumenting observations by drawing s,etches and ta,ing photographs or videos can be instructive and helpful for those who are not able to observe the real time dissolution test. 9bservations are especially useful during method development and formulation optimi.ation. "4amples of typical observations include, but are not limited to, the following5 (. %neven distribution of particles throughout the vessel. This can occur when particles cling to the sides of the vessel, when there is coning or mounding directly under the apparatus, when particles float at the surface of the medium, when film-coated tablets stic, to the vessel, andEor when off-center mounds are formed. 6. !ir bubbles on the inside of the vessel or on the apparatus or dosage unit. $heen on the apparatus is also a sign of air bubbles. This observation would typically be made when assessing the need to deaerate the medium. 7. ;ancing or spinning of the dosage unit, or the dosage unit being hit by the paddle. @. !dhesion of particles to the paddle or the inside of the bas,et, which may be observed upon removal of the stirring device at the end of the run. *. Pellicles or analogous formations, such as transparent sacs or rubbery, swollen masses surrounding the capsule contents. 2. Presence of large floating particles or chun,s of the dosage unit. >. 9bservation of the disintegration rate (e.g., percentage reduction in si.e of the dosage unit within a certain time frame#. 3. Aomple4 disintegration of the coating of modified or enteric-coated products<for e4ample, the partial opening and splitting apart (li,e a clamshell# or incomplete opening of the shell accompanied by the release of air bubbles and e4cipients. $ampling =anual< =anual sampling uses plastic or glass syringes, a stainless steel cannula that is usually curved to allow for vessel sampling, a filter, andEor a filter holder. The sampling site must conform to specifications under ;issolution >(( . !utosampling< !utosampling is a useful alternative to manual sampling, especially if the test includes several time points. -owever, because regulatory labs may perform the dissolution test using manual sampling, autosampling requires validation with manual sampling. There are many brands of autosamplers, including semiautomated and fully automated systems. 8outine performance chec,s, cleaning, and maintenance as described in the pertinent standard operating procedures or metrology documents are useful for reliable operation of these devices. $ome instruments are equipped with sampling through the bas,et or paddle shaft. Proper validation (e.g., demonstrated equivalence to results with the usual sampling procedure# may be required. The disturbance of the hydrodynamics of the vessel by sampling probes should be considered and adequate validation performed to ensure that the probes are not introducing a significant change in the dissolution rate. Aomparison of manual and automated procedures should be performed to evaluate the interchangeability of the procedures. This can be accomplished by comparing data from separate runs or, in some cases, by sampling both ways from the same vessel. 8esults should be consistent with the requirements for intermediate precision (described in this chapter in 'alidation# if the procedures are to be considered interchangeable.

9ther aspects of automation validation may include carryover of residual drug, effect of an in-residence probe (simultaneous sampling as mentioned above may not be suitable in this case#, adsorption of drug, and cleaning andEor rinse cycles. 0ilters 0iltration of the dissolution samples is usually necessary to prevent undissolved drug particles from entering the analytical sample and further dissolving. !lso, filtration removes insoluble e4cipients that may otherwise cause high bac,ground or turbidity. Prewetting of the filter with the medium may be necessary. 0ilters can be in-line or at the end of the sampling probe or both. The pore si.e can range from +.@* to >+ Rm. The usual types of filters are depth, dis,, and flow-through. -owever, if the e4cipient interference is high, if the filtrate has a cloudy appearance, or if the filter becomes clogged, an alternative type of filter or pore si.e should be evaluated. !dsorption of the drug(s# onto the filter needs to be evaluated. If drug adsorption occurs, the amount of initial filtrate discarded may need to be increased. If results are still unsuitable, an alternative filter material may be sought. 0ilter validation may be accomplished by preparing a suitable standard solution or a completely dissolved sample solution (e.g., prepared as a typical sample in a vessel or a sample put in a bea,er and stirred with a magnetic stirrer for ( hour#. 0or standard solutions, compare the results for filtered solutions (after discarding the appropriate volume# to those for the unfiltered solutions. 0or sample solutions, compare the results for filtered solutions (after discarding the appropriate volume# to those for centrifuged, unfiltered solutions. Aentrifugation Aentrifugation of samples is not preferred, because dissolution can continue to occur and because there may be a concentration gradient in the supernatant. ! possible e4ception might be for compounds that adsorb onto all common filters. !$$!H The usual assay for a dissolution sample is either spectrophotometric determination or -P:A. The preferred method of analysis is spectrophotometric determination because results can be obtained faster, the analysis is simpler, and fewer solvents are used. -P:A methods are used when there is significant interference from e4cipients or among drugs in the formulation to improve analytical sensitivity andEor when the analysis can be automated. It may be useful to obtain data for the drug with a stabilityindicating assay (e.g., -P:A chromatograms# in the medium of choice, even if the primary assay is based on a spectrophotometric method. '!:I;!TI9/ The validation topics described in this section are typical but not all-inclusive. The validation elements addressed may vary, depending on the phase of development or the intended use for the data. 2 The acceptance criteria are presented as guidelines only and may differ for some products. 0irms should document the appropriate acceptance criteria for their products in pertinent $9Ps. 9ther considerations may be important for special dosage forms. The e4tent of validation depends on the phase of the product development. 0ull validation ta,es place by the time of Phase III clinical studies. 'alidation studies should address the variations associated with different profile time points. 0or products containing more than a single active ingredient, the dissolution method needs to be validated for each active ingredient. $pecificityEPlacebo Interference It is necessary to demonstrate that the results are not unduly affected by placebo constituents, other active drugs, or degradates. The placebo consists of all the e4cipients and coatings (in,s, sin,er, and capsule shell are also included when appropriate# without the active ingredient. Placebo interference may be determined by weighing samples of the placebo blend and dissolving or dispersing them in dissolution medium at concentrations that would be encountered during testing. It may be desirable to perform this e4periment at 7> by comparing it to the (++D standard by the formula5 (++A(!P E !$#('E:# in which A is the concentration, in mg per m:, of the standardF !P and !$ are the absorbances of the placebo and the standard, respectivelyF ' is the volume, in m:, of the mediumF and : is the label claim, in mg. The interference should not e4ceed 6D.

note<0or e4tended-release products, a placebo version of the finished dosage form may be more appropriate to use than blends, because this placebo formulation will release the various e4cipients in a manner more nearly reflecting the product than will a simple blend of the e4cipients. In this case, it may be appropriate to evaluate potential interference at multiple sampling points in the release profile. If the placebo interference e4ceeds 6D, then method modification<such as ((# choosing another wavelength, (6# baseline subtraction using a longer wavelength, or (7# using -P:A<may be necessary in order to avoid the interference. Bhen other active drugs or significant levels of degradates are present, it is necessary to demonstrate that these do not significantly affect the results. 9ne procedure for doing this is to measure the matri4 in the presence and absence of the other active drug or degradate5 any interference should not e4ceed 6D. :inearity and 8ange :inearity and range are typically established by preparing solutions of the drug, ranging in concentration from below the lowest e4pected concentration to above the highest concentration during release. This may be done in conjunction with accuracyErecovery determination. The scheme may be altered if different flow-cell si.es or injection volumes are used. Typically, solutions are made from a common stoc, if possible. 0or the highest concentration, the determination may not e4ceed the linearity limits of the instrument. 9rganic solvents may be used to enhance drug solubility for the preparation of the standard solutionsF however, no more than *D (vEv# of organic solvent in the final solution should be used, unless validated. :inearity is typically calculated by using an appropriate least-squares regression program. Typically, a square of the correlation coefficient (r6 +.)3# demonstrates linearity. In addition, the y-intercept must not be significantly different from .ero. !ccuracyE8ecovery !ccuracyErecovery are typically established by preparing multiple samples containing the drug and any other constituents present in the dosage form (e.g., e4cipients, coating materials, capsule shell# ranging in concentration from below the lowest e4pected concentration to above the highest concentration during release. In cases of poor drug solubility, it may be appropriate to prepare a stoc, solution by dissolving the drug substance in a small amount of organic solvent (typically not e4ceeding *D# and diluting to the final concentration with dissolution medium. !n amount of stoc, solution equivalent to the targeted label claim may be added to the vessel instead of the drug powder. $imilarly, for very low strengths, it may be more appropriate to prepare a stoc, solution than to attempt to weigh very small amounts. The measured recovery is typically )*D to (+*D of the amount added. rac,eting or matri4ing of multiple strengths may be useful. ! special case for validation is the !cid $tage procedure described in ;elayed-8elease ;osage 0orms under ;issolution >((

. The limit of not more than (+D needs to be validated. If the compound degrades in acid, the validation e4periment must address this fact. Precision 8epeatability< 8epeatability is determined by replicate measurements of standard andEor sample solutions. It can be measured by calculating the 8$; of the multiple injections or spectrophotometric readings for each standard solution, or from the accuracy or linearity data. Intermediate Precision< Intermediate precision may be evaluated to determine the effects of random events on the precision of the analytical procedure. This evaluation is typically done later in the development of the drug product. The precision can be across the range of product strengths. Typical variations to study include days, analysts, and equipment. The use of an e4perimental matri4 design is encouraged for evaluation of intermediate precision. If possible, intermediate precision can be evaluated using a well-characteri.ed lot of drug product of tight content uniformity. In cases where a well-characteri.ed product is not available, placebo and active ingredient may be used to identify intermediate precision. The dissolution profiles on the same sample may be run by at least two different analysts, each analyst preparing the standard solutions and the medium. Typically, the analysts use different dissolution baths, spectrophotometers or -P:A equipment (including columns#, and autosamplersF and they perform the test on different days. This procedure may not need to be performed for each strengthF instead, brac,eting with high and low strengths may be acceptable.

! typical acceptance criterion is that the difference in the mean value between the dissolution results at any two conditions using the same strength does not e4ceed an absolute (+D at time points with less than 3*D dissolved and does not e4ceed *D for time points above 3*D. !cceptance criteria may be product-specific, and other statistical tests and limits may be used. 8obustness The evaluation of robustness, which assesses the effect of ma,ing small, deliberate changes to the dissolution conditions, typically is done later in the development of the drug product. The number of replicates (typically 7 or 2# is dependent on the intermediate precision. Parameters to be varied are dependent on the dissolution procedure and analysis type. They may include medium composition (e.g., buffer or surfactant concentration#, p-, volume, agitation rate, and temperature. 0or -P:A analysis, parameters may include mobile phase composition (percentage organic, buffer concentration, p-#, flow rate, wavelength, column temperature, and multiple columns (of the same type#. 0or spectrophotometric analysis, the wavelength may be varied. $tandard and $ample $olution $tability The standard solution is stored under conditions that ensure stability. The stability of the standard is analy.ed over a specified period of time, using a freshly prepared standard solution at each time interval for comparison. The acceptable range for standard solution stability is typically between )3D and (+6D. The sample solution is typically stored at room temperature. The sample is analy.ed over a specified period of time using the original sample solution response for comparison. The typical acceptable range for sample solution stability may be between )3D and (+6D compared with the initial analysis of the sample solutions. If the solution is not stable, aspects to consider could be temperature (refrigeration may be needed#, light protection, and container material (plastic or glass#. The procedure may state that the standards and samples need to be analy.ed within a time period demonstrating acceptable standard and sample solution stability. $pectrophotometric !nalysis $amples may be automatically introduced into the spectrophotometer using autosippers and flow cells. 8outine performance chec,s, cleaning, and maintenance as described in the standard operating procedures or metrology documents are useful for reliable operation of these instruments. Aells with path lengths ranging from +.+6 to ( cm are typically used. Aell alignment and air bubbles could be sources of error. The smaller path length cells are used to avoid diluting the sampleF however, acceptable linearity and standard error need to be demonstrated. ;uring analysis, standard solutions are typically prepared and analy.ed at just one concentration at (++D (or the selected C value# of the dosage strength. ;uring profile analysis, other concentrations may be useful. ! typical blan,, standard, and sample may be analy.ed in a sequence that brac,ets the sample with standards and blan,s, especially at the beginning and end of the analysis. In most cases, the mean absorbance of the dissolution medium blan, may not e4ceed (D of the standard. 'alues higher than (D must be evaluated on a case-by-case basis. The typical 8$; for %' analysis is usually not more than 6D. The absorptivity is calculated by dividing the mean standard absorbance by the concentration, in mg per m:, divided by the flow-cell path length in cm. !fter enough historical data are accumulated, an acceptable absorptivity range for the analyte (using the appropriate flow cell# may be determined. This value may be useful in troubleshooting aberrant data. 0iber optics as a sampling and determinative method, with proper validation, is an option. It may be useful to e4amine the %' spectrum of the drug in solution to select the optimum wavelength. -P:A 0or -P:A analysis, the compatibility of dissolution media and mobile phase may be e4amined, especially if large injector volumes (over (++ R:# are needed. $amples are normally analy.ed with -P:A using a spectrophotometric detector and an autoinjector. $ingle injections of each vessel time point with standards throughout the run constitute a typical run design. $ystem suitability tests include, at a minimum, the retention window and injection precision. Typically, the repeatability of an -P:A analysis should be less than or equal to 6D 8$; for five or si4 standard determinations. The standard level is typically at the (++D label claim level, especially for a single-point analysis. Preparation of the placebo samples for the -P:A analysis is to be performed in the same way as in the spectrophotometric analysis. "4amine the chromatogram for pea,s eluting at the same retention time as the drug. If there are e4traneous pea,s,

inject the standard solution, and compare retention times. If the retention times are too close, spi,e the placebo solution with the drug. Ahromatograms may also be obtained over an e4tended run time using the blan, (dissolution medium#, standard, and sample solution to identify late eluters that may interfere with subsequent analyses. The validation documentation may include overlaid representative chromatograms or spectra of blan, dissolution medium, a filtered placebo solution, a standard solution, and a filtered dissolution sample. !bsence of interfering pea,s in the placebo chromatogram or lac, of absorbance by the placebo at the analytical wavelength demonstrates specificity. !AA"PT!/A" A8IT"8I! Typical acceptance criteria for the amount of active ingredient dissolved, e4pressed as a percentage of the labeled content (C#, are in the range of >*D to 3+D dissolved. ! C value in e4cess of 3+D is not generally used, because allowance needs to be made for assay and content uniformity ranges. 3 !cceptance criteria including test times are usually established on the basis of an evaluation of the dissolution profile data. !cceptance criteria should be consistent with historical data, and there is an e4pectation that acceptable batches (e.g., no significant differences in in vivo performance, composition, or manufacturing procedure# will have results that fall within the acceptance criteria. ( The iopharmaceutics Alassification $ystem is outlined in the 0;! ?uidance for Industry5 Baiver of In 'ivo ioavailability and ioequivalence $tudies for Immediate-8elease $olid 9ral ;osage 0orms ased on a iopharmaceutics Alassification $ystem, !ugust 6+++F http5EEwww.fda.govEcderEguidanceE72(3fnl.htm, accessed 2E66E6++*. 6 oudreau, $.P.F =c"lvain, 1.$.F =artin, :.;.F ;owling, T.F 0ields, $.=. =ethod 'alidation by Phase of ;evelopment, an !cceptable !nalytical Practice. Pharmaceutical Technology 6++@F 63(((#5*@L22. 7 $ee the 0;! ?uidance for Industry5 ;issolution Testing of Immediate-8elease $olid 9ral ;osage 0orms, !ugust ())>F http5EEwww.fda.govEcderEguidanceE(>(7bp(.pdf, accessed 2E66E6++*.

Vous aimerez peut-être aussi