A Strategy For Quality Control Dissolution Method .

3y ago
35 Views
3 Downloads
527.75 KB
7 Pages
Last View : 8d ago
Last Download : 3m ago
Upload by : Lucca Devoe
Transcription

dx.doi.org/10.14227/DT220315P10A Strategy for Quality Control DissolutionMethod Development for Immediate-ReleaseSolid Oral Dosage FormsGerard M. Bredael1,*, Steve Liang2, and David Hahn31 PharmaceuticalSciences and Clinical Supply, Preformulation, Merck, Summit, NJ USAChemistry in Development and Supply, Merck, Summit, NJ USA3 Formerly from Pharmaceutical Sciences and Clinical Supply, Analytical Sciences, Merck, Summit, NJ USA2 Analyticale-mail: gerard.bredael@merck.comABSTRACTDevelopment of an appropriate dissolution method for quality control purposes requires consideration of many factorsand a balance of different needs. The method is typically distinctive to each drug product because of the uniquecharacteristics of the active ingredient, the formulation, and the manufacturing process. The method developmentand selection process remains subjective due to the potential range of profiles that could be considered acceptable.This article presents a strategy and detailed recommendations for developing a quality control dissolution method forimmediate-release solid oral dosage forms for the purpose of quality control and formulation or process development.The strategy is developed based on an understanding of dissolution technology, regulatory expectations, literature, andexperiences in developing various dissolution methods. A case study demonstrating this strategy with the discriminatorypotential of an appropriately developed dissolution method is also described.KEYWORDS: Dissolution; oral drug delivery; formulation; solubility; in vitro models.INTRODUCTIONDissolution testing of solid oral dosage forms hasbeen used for several decades to aid in formulation/process development and to examine and assurebatch-to-batch quality, consistency, and performance ofdrug products (1). The USP General Chapter 1092 TheDissolution Procedure: Development and Validation hasuseful explanations on the development aspects. USPGeneral Chapter 711 Dissolution has the specifics fordissolution testing methodology. There are articles thatprovide more dissolution development background. Skouget al. (2) gave one of the first comprehensive overviewsof the development, validation, and specification settingfor dissolution testing. The importance of a compound’sbiopharmaceutical aspects and a dissolution testdesign–decision tree based on the BiopharmaceuticsClassification System (BCS) was presented by Li et al. (3).Lastly, Gray et al. (4) provided a historical perspective, thebroad challenges of development, and applications.While these resources provide a broad foundation fordissolution method development, this article presentsa simple step-by-step strategy for developing a qualitycontrol dissolution method for immediate-releasesolid oral dosage forms. Specific guidance and detailedrecommendations are provided. Relevant articles arecited as background for some of the dissolution conditionsand development considerations.10*Corresponding author.Dissolution Technologies AUGUST 2015MATERIALS AND METHODSMaterialsThe compound examined in the case study is a salt form ofa weak base. Wet-granulated immediate-release tabletsof this compound were used. The dissolution bufferswere prepared from analytical grade reagents.MethodsThe dissolution testing was performed on Distek 2100Cdissolution baths. The paddle method was used withvarious media. The dissolution analyses were performedin situ with a LEAP Technologies OPT DISS UV fiber-opticsystem. The 10-mm probes were used with 320-nmwavelength detection.DISSOLUTION TEST DEVELOPMENT GUIDEA dissolution method development process is describedbelow. Each important parameter of a dissolutiontest is separated into individual sections to allow easyidentification. The strategy itself was created aroundhealth authority guidances or guidelines. This guidepresents aspects of dissolution method development forultimately creating a method acceptable to regulatoryagencies.Solubility Based on BCSThe most important data set for dissolution methoddevelopment is the solubility–pH profile. The solubilityprofile will indicate whether the compound is considereda highly soluble compound based on the BCS. If the

highest proposed strength dosage dissolves in 250 mLof media over the pH range 1–6.8 according to the EMAguidance (5) or pH 1–7.5 according to the United StatesFDA guidance (6), then it is considered a highly solublecompound.If the compound is highly soluble, dissolution profilesshould be established using 900 mL of 0.1 N HCl, pH 4.5,and pH 6.8 media, with typically USP Apparatus 2 (paddles)at 50 rpm. The medium that produces the slowestdissolution rate with a standard spindle speed should beselected for the method. A slower dissolution rate willincrease the likelihood that the method might be ableto discriminate formulation composition, manufacturingprocess variations, or pharmacokinetics performance.Lastly, the appropriate medium selection would makethe method suitable for determining whether the dosageform still would meet BCS Class 3, Class 1, or both criteriafor a highly soluble compound over the shelf-life.If the compound has low solubility across the pH range,then the initial objective is to develop a dissolutionmethod that dissolves at least 85% by 30–60 min (7).Additionally, if an animal PK study has been performedusing different API particle sizes, different physical forms,or other critical attributes and PK differences are observed,the method ideally should be capable of rank-orderingthe formulations. Subsequent sections provide guidanceon how to approach development of an appropriatedissolution test for low-solubility compounds.ApparatusFor immediate-release solid oral dosage forms, USPApparatus 1 (Basket) or Apparatus 2 (paddle) are typicallyused. The other USP dissolution apparatus are typicallyused for controlled-release or non-oral formulations. Thepaddle apparatus should be chosen if the anticipatedcommercial dosage form will be a non-floating dosageform, unless there are extenuating circumstances. Thepotential issue with baskets for disintegrating formulationsis that the hydrodynamic environment below a basket isnot as well mixed as that of the paddle, which may leadto a more challenging interpretation of the dissolutiondata. Sometimes the dosage form used in an early phasemay be different from the final commercial dosage form.If an early dosage form (e.g., capsules) floats, the useof a sinker around the capsule should be considered toallow a paddle method to be used. This will permit theeasiest tracking of dissolution data for the conversionfrom capsule to tablet formulation with the removal ofthe sinker in the test. It is a good regulatory strategy tominimize dissolution method changes across all phases.For a floating dosage form, baskets and paddles withsinkers should be evaluated. In some cases, basketsmay provide an advantage for nondisintegrating dosageforms. Baskets would create the same hydrodynamicenvironment reproducibly for these dosage forms as wellas ensure that the medium has free access to the dosageform.Spindle SpeedWith the paddle apparatus, a 50-rpm spindle speedshould be used as the starting point based on regulatoryguidances from FDA (7), the European Medicines Agency(EMA) (5), and the Japanese Pharmaceutical and FoodSafety Bureau (PFSB) (8). If there are issues with coning(the piling of non-dissolving excipients under thepaddle that limits media penetration into the pile), theuse of paddles with a 75-rpm spindle speed should beinvestigated. The FDA and PFSB recommend a 75-rpmpaddle as an option. The increased paddle speed maydisperse excipients better, mimicking in vivo dispersion,and allow unhampered dissolution. A 100-rpm paddlemethod may be used with sufficient justification, such aseliminating or reducing surfactant concentration.With the basket apparatus, a spindle speed of 100 rpmshould be investigated initially (5, 7, 8). The FDA (7) alsoadds the option of baskets at 50 rpm. If dissolution is toorapid to provide a potentially discriminating profile, the50-rpm basket can be investigated.Use of spindle speeds other than those recommendedin regulatory guidance documents should be consideredonly when recommended parameters have beenexhausted. Use of alternative speeds should be clearlyjustified. Selecting a different medium (e.g., one that hasa higher compound solubility) to accelerate dissolution ispreferred to increasing the spindle speed.A good diagnostic tool for development is an “infinity spin”added to the end of a dissolution method to try to forciblybreak apart granules and dissolve any undissolved activepharmaceutical ingredient (API). After the last normalsampling time point, the spindle speed is increased to150–250 rpm for 15–30 min, and an additional sampleis taken. This can provide a quick check on dosage formpotency and assure that the dissolution is not solubilitylimited or that a low dissolution is not due to low potency.However for batch release testing, the added value of aninfinity spin is limited.Media and BuffersDissolution testing with biorelevant media may be usefulfor internal decision-making purposes during formulationdevelopment; however, the QC test could use a completelyDissolution Technologies AUGUST 201511

separate test method (9). Biorelevant media are designedto mimic the complexity of human GI tract solutionsand are frequently used during development to betterunderstand a compound’s potential in vivo solubilityand stability and for formulation screening. JenniferDressman at the Johann Wolfgang Goethe University hasbeen involved in researching the complexity of humanGI solutions and has published multiple articles (10–15)on creating biorelevant solutions with subsequentevaluations using these solutions. Note that methodsusing biorelevant media are not necessarily biopredictive(linked to a compound’s clinical behavior) unless suchrelationships have been established with clinical studydata. QC testing utilizing these biorelevant solutions,however, would be cost-prohibitive, resource demanding,and complex (potential for error). The use of these mediaincreases the difficulty of developing a robust analyticalmethod for QC purposes. For these reasons, simple buffersystems are preferred for routine dissolution analysis.Depending on the compound, such methods may havethe potential to be biopredictive.The type of medium and the volume are selected toprovide sink conditions. USP defines sink condition as“the volume of medium at least three times that requiredin order to form a saturated solution of drug substance.”The solubility-versus-pH profile provides the most usefulinformation in medium selection for initial examination.Skoug et al. (2) stated that in vitro in vivo correlations aremore likely obtained near the saturated solubility limit.However, as the dissolution method approaches thissolubility limit, the risk that the test may be oversensitivealso increases while the robustness of the method tendsto decrease. Initially selecting a dissolution methodwith a relatively slow dissolution rate that is still not lessthan 85% dissolved by 60 min could provide sufficientdiscriminatory capability and therefore minimize the riskthat a more sensitive test late in the development stageis needed to demonstrate sensitivity to formulation orprocess changes. Switching to a more discriminatorydissolution method late in development could potentiallyincrease the criticality or encumbrance of such a change.12The general pH range of dissolution media is from 1.1to 6.8. The pH can be higher if needed for solubilityreasons. In general, the pH should not exceed 8.0 (7). Amedium is chosen based on the desired pH, for example,hydrochloric acid for pH 1.0–3.0, glycine for pH 2.0–3.0, citrate for pH 2.5–3.5, acetate for pH 4.0–5.5, andphosphate pH 6.0–8.0. These stated buffer pH rangesare by no means limitations. A typical dissolution bufferhas a 0.05 molar concentration. Unbuffered water is nota preferred medium due to potential variability in pHdepending on the source.Dissolution Technologies AUGUST 2015If the medium over the pH range fails to give adequatedissolution, then surfactants should be evaluated. Thefirst choice for a surfactant is sodium dodecyl sulfate(SDS) that is also called sodium lauryl sulfate (SLS) (7).SDS is the most commonly used surfactant in dissolutionmedia because it is available in high purity withconsistency across vendors and is easy to use for accurateconcentration preparation. SDS tends to degrade belowpH 2 but has successfully been used in such media (16).For these cases, a stability or use period should beprovided. A pure form of SDS of at least 98% purity shouldbe specified. Previously accepted levels of SDS and othersurfactants used in a regulatory filing can be found on theFDA dissolution Web site (16).The concentration of the surfactant used for dissolutiontest should be justified. Typically the lowest concentrationrequired to achieve an acceptable dissolution profileshould be used. An SDS concentration range of 0.1–3%or higher has been used (16). In unbuffered water, SDSat a concentration below about 0.23% behaves moreas a wetting agent than as a solubilizing agent becauseit is below its critical micelle concentration. If SDS at apreviously approved concentration does not yield at least85% dissolved by 60 min or interacts with the active/excipients, other surfactants listed on this FDA Web site(16) should be examined.The effect of medium deaeration should be investigated.Room-temperature medium can hold more dissolvedgases than medium at 37 C. When the medium is heated,the dissolved gases have a tendency to form bubbles.These bubbles could have an unpredictable effect ondissolution. The bubbles may cause a dosage form toadhere to the apparatus/vessel, reduce medium access toparticles, or increase the tendency for particles to float.A significant effect could be observed in variability or achange in the dissolution rate.Medium VolumeThe standard dissolution medium volumes used in theindustry and accepted by regulatory agencies are 500 mLand 900 mL. These volumes are selected to provide sinkconditions for the compound and do not represent thevolumes of liquid encountered by the product in vivo. Indepth reviews of the human gastrointestinal physiologyare documented by McConnell and Mudie (17, 18).Smaller dissolution medium volumes can be used duringdevelopment to reduce API supply requirements, andlarger volumes up to 4 L may be used if required forsink–solubility reasons. In either case, an appropriateapparatus is required. The dissolution behavior (variabilityand profile) of the dosage form itself will be the best

guide in choosing the volume. An effort to use one ofthe two standard volumes should be made to facilitatemethod transfer and reduce the likelihood of regulatoryquestions.It is preferred to have one method and therefore onevolume across all strengths of a compound. This allowsthe evaluation of the profiles across strengths and a checkon the dissolution of same/similar formulations. Such amethod shows whether the higher and lower strengthsare dissolving at similar rates in vitro. Additionally, use ofthe same method across dosage strengths may provideopportunities for bracket-testing of only the high and lowstrengths under certain circumstances.Sampling Time PointsDuring development of IR products, sampling time pointsgenerally range from 5 min to more than 60 min. The5-min time point may be used for suspensions or other fastdissolving formulations where the variation is not high.Typical time points are 15, 30, 45, and 60 min. However,the time points are based on the product’s profile andon the method’s ability of tracking key aspects of theformulation. If there is a desire to better understand ortrack the disintegration effect, a 5- or 10-min time pointwould be needed. (For fast-dissolving products, a fiberoptic dissolution system could test more time points toobtain a better defined dissolution profile.) If there is asignificant risk of slowing on stability, a time point maybe added beyond 60 min to ensure that at least 85%dissolved average values are achieved.It is useful to have a sampling time point at 15 min,especially if the compound is dissolved at least 85% withinthat period of time. If this is achieved in 0.1 N HCl, the FDA(7) considers that the dosage “behaves like a solution andgenerally should not have any bioavailability problems.”.It is also the specified time point and specification fora potential BCS 3 biowaiver (5). Finally, in comparingdissolution profiles, an f2 calculation, which is a logarithmictransformation of the sum-squared error differencesbetween the dissolution curves, is not required forsimilarity justification with at least 85% dissolved (6) orgreater than 85% dissolved (5) in 15 min.SinkersAs mentioned previously, sinkers can be utilized oncapsules to allow use of the USP 2 Apparatus. Sinkersmay also aid in other situations, such as sticky tablets orslowly disintegrating tablets. Tablets sticking to vesselsmay result in high variability in dissolution profilesbecause they may stick at various off-center locationsin the vessels. The non-centered tablets are exposed toa different hydrodynamic environment than those thatare centered (19, 20). Slowly disintegrating tablets mayneed fluid flowing around the tablets to generate moreconsistent dissolution profiles. Placing the dosage formsin sinkers may resolve these concerns, allowing the useof the paddle apparatus. Various sinker configurationsand models should be tested to find one that gives thedesired results. Lastly, the sinker configuration or modelshould be specified in the method due to the potentialeffects that different sinker models may have on thehydrodynamics surrounding the dosage form.Filtration and Endpoint AnalysisFiltration of dissolution samples should eliminatepost-sampling dissolution of API particles andreduce the potential that excipient particles mightcreate backpressure/clogging issues in the analyticalinstrumentation. Typical filter pore sizes range from 0.45to 70 μm. For micronized drug substance, the analystshould strive to utilize the filter with the smallest feasiblepore size .The analytical method will depend on the dosage form,amount of compound, and compound UV absorptivity.UV analysis using a fiber-optic or online instrument isthe most efficient technique available for dissolutionsample analysis, when applicable. Also, online UV orfiber-optic analysis will provide the dissolution results atthe end of the dissolution test for fast turnaround time.Applicability of UV methods depends on both suitableabsorptivity and absence of significant capsule shell orexcipient interference. HPLC/UPLC analysis is the othertypical method. With regard to HPLC, it is better to injectlarger volumes to maintain sensitivity for lower strengthdosages than to decrease the dissolution medium volumeto increase HPLC response.RobustnessThe effect of the various dissolution parameters (pH,surfactant concentration, etc.) on the dissolutionprofiles should be mapped to assess robustness duringdevelopment. For example, the dissolution robustnesswith regard to minor changes in the pH should be checked.A deviation of 0.05 pH units should have no significantimpact on the dissolution rate. If there is, a different pHshould be chosen for robustness reasons.Evaluation of Method Sensitivity/DiscriminatoryPowerBatches prepared from variants of the target commercialformulation levels or processing parameters aretypically used to analyze the sensitivity or discriminatorypower of the dissolution method. Among the factorsfor consideration are drug substance particle size,formulation composition (such as low disintegrant), and aprocess variation (such as over-granulation). The variantsDissolution Technologies AUGUST 201513

Subsequent DevelopmentTable 1 is a summary of the key recommendationsfor developing an early phase dissolution method

process development and to examine and assure batch-to-batch quality, consistency, and performance of drug products (1). The USP General Chapter 1092 The Dissolution Procedure: Development and Validation has useful explanations on the development aspects. USP General Chapter 711 Dissolution has the specifics for dissolution testing methodology.

Related Documents:

Bruksanvisning för bilstereo . Bruksanvisning for bilstereo . Instrukcja obsługi samochodowego odtwarzacza stereo . Operating Instructions for Car Stereo . 610-104 . SV . Bruksanvisning i original

10 tips och tricks för att lyckas med ert sap-projekt 20 SAPSANYTT 2/2015 De flesta projektledare känner säkert till Cobb’s paradox. Martin Cobb verkade som CIO för sekretariatet för Treasury Board of Canada 1995 då han ställde frågan

service i Norge och Finland drivs inom ramen för ett enskilt företag (NRK. 1 och Yleisradio), fin ns det i Sverige tre: Ett för tv (Sveriges Television , SVT ), ett för radio (Sveriges Radio , SR ) och ett för utbildnings program (Sveriges Utbildningsradio, UR, vilket till följd av sin begränsade storlek inte återfinns bland de 25 största

Hotell För hotell anges de tre klasserna A/B, C och D. Det betyder att den "normala" standarden C är acceptabel men att motiven för en högre standard är starka. Ljudklass C motsvarar de tidigare normkraven för hotell, ljudklass A/B motsvarar kraven för moderna hotell med hög standard och ljudklass D kan användas vid

LÄS NOGGRANT FÖLJANDE VILLKOR FÖR APPLE DEVELOPER PROGRAM LICENCE . Apple Developer Program License Agreement Syfte Du vill använda Apple-mjukvara (enligt definitionen nedan) för att utveckla en eller flera Applikationer (enligt definitionen nedan) för Apple-märkta produkter. . Applikationer som utvecklas för iOS-produkter, Apple .

This presentation and SAP's strategy and possible future developments are subject to change and may be changed by SAP at any time for any reason without notice. This document is 7 provided without a warranty of any kind, either express or implied, including but not limited to, the implied warranties of merchantability, fitness for a .

och krav. Maskinerna skriver ut upp till fyra tum breda etiketter med direkt termoteknik och termotransferteknik och är lämpliga för en lång rad användningsområden på vertikala marknader. TD-seriens professionella etikettskrivare för . skrivbordet. Brothers nya avancerade 4-tums etikettskrivare för skrivbordet är effektiva och enkla att

Den kanadensiska språkvetaren Jim Cummins har visat i sin forskning från år 1979 att det kan ta 1 till 3 år för att lära sig ett vardagsspråk och mellan 5 till 7 år för att behärska ett akademiskt språk.4 Han införde två begrepp för att beskriva elevernas språkliga kompetens: BI