active compounds release from semisolid dosage forms

14
7/21/2019 Active Compounds Release From Semisolid Dosage Forms http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 1/14 REVIEWS Active Compounds Release from Semisolid Dosage Forms ANNA OLEJNIK, JOANNA GOSCIANSKA, IZABELA NOWAK Adam Mickiewicz University in Poznan, Faculty of Chemistry, ul. Umultowska 89b, 61-714 Pozna´ n, Poland  Received 20 June 2012; revised 17 July 2012; accepted 18 July 2012  Published online 8 August 2012 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23289  ABSTRACT:  The aim of this paper is to review all the aspects of the  in vitro  release test- ing (IVRT) from semisolid dosage forms. Although none of the official dissolution methods has been specified for use with semisolid dosage forms, their utility for assessing release rates of drugs from semisolid dosage forms has become a topic of considerable interest. One can expect to overcome such complexity in the future, when the official “Topical and Transdermal Drug Products—Product Performance Tests” will be published in an issue of the Pharmacopeial Fo- rum. Many factors such as type of the dissolution medium, membrane, temperature, and speed have an influence on the mechanism and kinetics of the release testing from gels, creams, and ointments; therefore, those parameters have been widely discussed. © 2012 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 101:4032–4045, 2012 Keywords:  active transport;controlledrelease; diffusion;emulsions; formulation; gels; kinet- ics;  in vitro models INTRODUCTION In pharmaceutical industry, dissolution testing was primarily used as a fundamental tool in quality control of the solid oral dosage forms including immediate-/sustained-action tablets and capsules. 1–3 However, this method was extended to analyze semisolid dosage forms (such as creams, gels, and ointments) as well. The method is known as  in vitro release testing (IVRT) because the active ingredient has to diffuse and be released by the vehicle. 4–7 Semisolid dosage forms cause distinctive problems in the development of  in vitro  release technologies simply because of the physicochemical properties of formulations and the specific physiological environ- ment in which they should release their content. 8–11 Those products are complex formulations, which are composed of two phases (water and oil), one is an external phase and the other is an internal phase (Fig. 1). 12,13 The active compound is usually dissolved in one of the above-mentioned phases. However, oc- casionally, the active compound is not fully soluble; therefore, a three-phase system is created because of the dispersing of the active compound in one or both of the phases. Diverse factors such as the size of the dis- persed particles, the interfacial tension between the Correspondenceto: Anna Olejnik(Telephone: +48-61-829-12-95; Fax: +48-61-829-12-07; E-mail: [email protected]) Journal of Pharmaceutical Sciences, Vol. 101, 4032–4045 (2012)  © 2012 Wiley Periodicals, Inc. and the American Pharmacists Association phases, andtherheologyoftheformulationdetermine the physical properties of the semisolid dosage form.  All of these factors have an influence on the release profile of the active ingredient. 4,8,9,11,13  In vitro  release testing is recommended by the United States Food and Drug Administration as a measure of “product sameness” during scale-up and postapproval changes for semisolids (SUPAC- SS). This method is routinely used during product development to fine-tune a formulation. However, the suggested uses of the tests to establish batch- to-batch uniformity, product certification, and possi- ble bioequivalence are fairly new and have been the subject of much debate. It is important to properly  validate a release test before using it for product qualification. 4,6,15–17 It must be reproducible and re- liable, and although it is not a measure of bioavail- ability, the performance test must be capable of de- tecting changes in drug-release characteristics from the finished product. Changes in drug-release char- acteristics have the potential to alter the biological performance of the drug in the dosage form. Such changes may be related to active or inactive/inert in- gredients in the formulation, physical or chemical at- tributes of the finished formulation, manufacturing  variables, shipping and storage effects, aging effects, and other formulation factors that are critical to the quality characteristics of the finished drug product.  An abundant literature exists on in vitro release of drugs present in suspension semisolid formulations. 7 4032  JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Upload: sameh-ibrahim-qanadilo

Post on 04-Mar-2016

48 views

Category:

Documents


7 download

DESCRIPTION

The aim of this paper is to review all the aspects of the in vitro release testing(IVRT) from semisolid dosage forms. Although none of the official dissolution methods hasbeen specified for use with semisolid dosage forms, their utility for assessing release rates ofdrugs from semisolid dosage forms has become a topic of considerable interest. One can expectto overcome such complexity in the future, when the official “Topical and Transdermal DrugProducts—Product Performance Tests” will be published in an issue of the Pharmacopeial Forum.Many factors such as type of the dissolution medium, membrane, temperature, and speedhave an influence on the mechanism and kinetics of the release testing from gels, creams, andointments; therefore, those parameters have been widely discussed

TRANSCRIPT

Page 1: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 1/14

REVIEWS

Active Compounds Release from Semisolid Dosage Forms

ANNA OLEJNIK, JOANNA GOSCIANSKA, IZABELA NOWAKAdam Mickiewicz University in Poznan, Faculty of Chemistry, ul. Umultowska 89b, 61-714 Poznan, Poland

 Received 20 June 2012; revised 17 July 2012; accepted 18 July 2012

 Published online 8 August 2012 in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.23289

 ABSTRACT:   The aim of this paper is to review all the aspects of the   in vitro   release test-

ing (IVRT) from semisolid dosage forms. Although none of the official dissolution methods has

been specified for use with semisolid dosage forms, their utility for assessing release rates of 

drugs from semisolid dosage forms has become a topic of considerable interest. One can expect

to overcome such complexity in the future, when the official “Topical and Transdermal Drug 

Products—Product Performance Tests” will be published in an issue of the Pharmacopeial Fo-

rum. Many factors such as type of the dissolution medium, membrane, temperature, and speedhave an influence on the mechanism and kinetics of the release testing from gels, creams, and

ointments; therefore, those parameters have been widely discussed. © 2012 Wiley Periodicals,

Inc. and the American Pharmacists Association J Pharm Sci 101:4032–4045, 2012

Keywords:   active transport; controlled release; diffusion; emulsions; formulation; gels; kinet-

ics; in vitro models

INTRODUCTION

In pharmaceutical industry, dissolution testing wasprimarily used as a fundamental tool in qualitycontrol of the solid oral dosage forms including immediate-/sustained-action tablets and capsules.1–3

However, this method was extended to analyzesemisolid dosage forms (such as creams, gels, andointments) as well. The method is known as  in vitro

release testing (IVRT) because the active ingredienthas to diffuse and be released by the vehicle.4–7

Semisolid dosage forms cause distinctive problemsin the development of   in vitro   release technologiessimply because of the physicochemical properties of formulations and the specific physiological environ-ment in which they should release their content.8–11

Those products are complex formulations, which arecomposed of two phases (water and oil), one is an

external phase and the other is an internal phase(Fig. 1).12,13 The active compound is usually dissolvedin one of the above-mentioned phases. However, oc-casionally, the active compound is not fully soluble;therefore, a three-phase system is created because of the dispersing of the active compound in one or both of the phases. Diverse factors such as the size of the dis-persed particles, the interfacial tension between the

Correspondence to: Anna Olejnik (Telephone:+48-61-829-12-95;Fax: +48-61-829-12-07; E-mail: [email protected])

Journal of Pharmaceutical Sciences, Vol. 101, 4032–4045 (2012)

 © 2012 Wiley Periodicals, Inc. and the American Pharmacists Association

phases, and the rheology of the formulation determinethe physical properties of the semisolid dosage form.

 All of these factors have an influence on the releaseprofile of the active ingredient.4,8,9,11,13

 In vitro   release testing is recommended by the

United States Food and Drug Administration asa measure of “product sameness” during scale-upand postapproval changes for semisolids (SUPAC-SS). This method is routinely used during productdevelopment to fine-tune a formulation. However,the suggested uses of the tests to establish batch-to-batch uniformity, product certification, and possi-ble bioequivalence are fairly new and have been thesubject of much debate. It is important to properly

 validate a release test before using it for productqualification.4,6,15–17 It must be reproducible and re-liable, and although it is not a measure of bioavail-

ability, the performance test must be capable of de-tecting changes in drug-release characteristics fromthe finished product. Changes in drug-release char-acteristics have the potential to alter the biologicalperformance of the drug in the dosage form. Suchchanges may be related to active or inactive/inert in-gredients in the formulation, physical or chemical at-tributes of the finished formulation, manufacturing 

 variables, shipping and storage effects, aging effects,and other formulation factors that are critical to thequality characteristics of the finished drug product.

 An abundant literature exists on in vitro release of drugs present in suspension semisolid formulations.7

4032   JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 2: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 2/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4033

Figure 1.   The basic constituents of various semisolid dosage forms (ointments, creams, and

gels).14

The first theoretical description of release from dis-persions in ointment bases was published by Higuchiin 1961.18  A key assumption in this derivation wasthat the drug diffuses into a perfect sink. The matrix-boundary layer model was later developed to accountfor the resistance of a diffusion layer at the surfaceof a matrix.19 Tojo20 described a graphical method forobtaining the intrinsic release, defined as being in-

dependent of diffusion-layer contributions, from realdata that can include such contributions. The matrix-boundary layer model was used by Bottari et al.21 in1977 for an analysis of benzocaine release from gelsthrough an inert membrane into a receptor fluid.

Drug-release measurements from creams, gels, andointments, in connection with research on topical de-livery systems, have appeared in the literature formany years. Previously, the semisolid was placed indirect contact with a receptor liquid. A variation of this technique utilized an apparatus in which a screenloaded with a semisolid was lowered into the recep-

tor fluid. It is known that with these methods it is very important to guard against dissolution or dis-persion of the semisolid into the receptor. One can ex-pect the appearance of a physical disturbance of thesemisolid surface by the agitation needed to main-tain homogeneity within the receptor. Application of a membrane between the semisolid and the recep-tor was designed to prevent these artifacts.7 More-over, transport through the porous membrane is muchmore rapid than through the semisolid, but the mem-brane contributes to the overall diffusional resistance.

Until now, release testings of various types of ac-tive components from semisolid dosage forms were

Table 1.   Examples of Active Compounds Applied in Release

Studies (w/o - water-in-oil, o/w - oil-in-water,

o/w/o-oil-in-water-in-oil)

 Active Compound Semisolid Dosage Form References

Caffeine W/O emulsions, gels 22–25

Hydrocortisone Creams, ointment, lotions,

O/W, and O/W/O

emulsions

26–31

 Acyclovir Creams 32Retinol O/W/O emulsions 33

Retinoic acid Creams 34,35

Ketoprofen Ointments, gels 36–38

Chlorpheniramine

maleate

Ointments, gels 39–41

 Vitamin B12   Gels 42

Salicylic acid Ointments 43

Benzocaine Gels 21

Corticoids Ointments, creams 44,45

Phenol Ointments 46

Peptides Gels 47

Naproxen Ointments 48,49

Metronidazole W/O/W emulsions 50

Ibuprofen Ointments 51–53

Flurbiprofen Gels 54–56Heparin Gels 57–60

extensively studied and are presented in Table 1. Themain purpose of this article is to review all the aspectsof conventional and nonconventional IVRT methodsfor novel semisolid dosage forms. This review pro-

 vides insight into the possible alternatives for active-component-release study design and the choices of dissolution medium, membrane, temperature, as wellas the speed for such tests.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 3: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 3/14

4034   OLEJNIK, GOSCIANSKA, AND NOWAK 

METHODOLOGY OF IVRT

In 2009, two new draft United States Pharmacopeia(USP) general chapters on topical and transdermaldrug products were published in the  Pharmacopeial

 Forum ( PF ) 35(3), May–June 2009. The general chap-ters are   <3>Topical and Transdermal Products—

 Product Quality Tests and  <725>Topical and Trans-

dermal Products—Product Performance Tests. Thispublication was followed by a 2-day workshop “USPWorkshop on Topical and Transdermal Drug Prod-ucts” held in USP Headquarters from 14 to 15September, 2009. In 2010, a revision to the proposednew general test chapter was published on the basisof the comments received during the public commentperiod and at the above-mentioned workshop. How-ever, one has to be aware that a new general chapter,<1724>Topical and Transdermal Drug Products—

 Product Performance Tests, is under development andwill be published in a future issue of   PF . After its

publication, a cross-reference to the new chapter willbe added to the general chapter  <3>. As there areno effective directives in the pharmacopeia regard-ing the release system that should be employed, re-searchers have, therefore, designed different appara-tus and methods to carry out these tests. However,several considerations must be taken into account toobtain satisfactory, reliable, and repeatable results.The most important requirement is to choose the cor-rect quantities of the active ingredient and the othercomponents.61 The appropriate membrane, receptor,and temperature for the release testing must also be

selected.

Selection of an Appropriate Membrane

The membranes used in   in vitro   release studieswere developed to provide a physical support andmaintain constant contact between the formulationand receiving medium.62 There are some impor-tant considerations that must be taken into ac-count when selecting a membrane for release stud-ies. The membranes selected for   in vitro   diffusionstudies should be commercially available, inert, and

should not exhibit any physical or chemical interac-tion with the formulation.6,62 Moreover, the mem-brane should be compatible with the receptor andoffer the least-possible resistance to the diffusion of the active compound.7,63 The synthetic membraneshave been widely used to determine the   in vitro  re-lease rate of an active compound from various top-ical formulations.16,24,27,64,65 The commercial avail-ability, stability, and ease of use make synthetic mem-branes highly desirable. In addition, they exhibit theadded benefit of ensuring batch-to-batch homogeneityand uniformity, which is often lacking with biologicalmembranes.

Companies, involved in the field of transdermal de-livery, concentrate on a few selective polymeric sys-tems. For example, Alza Corporation (Mountain View,California) mainly concentrates on ethylene vinyl ac-etate (EVA) copolymers or microporous polypropy-lene, and Searle Pharmacia (Peapack, New Jersey)concentrates on silicone rubber.66 Similarly, Color-

con (Dartford, Kent, UK) uses hydroxypropyl methyl-cellulose (HPMC) for matrix preparation for propra-nolol transdermal delivery, and Sigma (St. Louis, Mis-souri) uses ethylcellulose for an isosorbide dinitratematrix.67–69 The polymers utilized for transdermaldrug delivery system (TDDS) can be classified as:

•   natural polymers, for example, cellulose deriva-tives, gelatin, waxes, gums, natural rubber, chi-tosan, and so on70;

•   synthetic elastomers, for example, polybutadi-ene, polyisobutylene, silicone rubber, silastic

membrane (polydimethylsiloxane), nitrile, acry-lonitrile, and so on;

•   synthetic polymers, for example, polyvinyl alco-hol, polyvinylchloride, polyethylene, polypropy-lene, polyacrylate, polyamide, polyurea,polyvinylpyrrolidone, polyvinylidene difluo-ride, polymethylmethacrylate, and so on.

In order to form a TDDS matrix, different polymers(such as cross-linked polyethylene glycol, eudragits,ethylcellulose, polyvinylpyrrolidone, and HPMC) areused. Other polymers such as EVA,71 silicone rub-

ber, and polyurethane72 are widely employed as rate-controlling membranes. Commonly used membranesin  in vitro  release tests (Table 2) are fluorophore hy-drophobic membrane, thermoplastic silicone polycar-bonate urethane (Carbosil R, DSM Biomedical, Berke-ley, CA), polysulfone, cellulose acetate, polytetraflu-oroethylene, spectrapore dialysis membrane, mixedesters of cellulose and natural skin (human,31,73

rat,45,68,70 rabbit, and mouse),31 reconstituted humanepidermis,74,75 Nylon, Teflon, and polycarbonate.2

The use of porous membranes facilitates the diffu-sion of drugs. The barrier potential of porous mem-

branes is determined by the possibility for a moleculeto enter and diffuse through the pores. This possi-bility is, in turn, affected by factors such as the rela-tive molecular size, molecular shape, and electrostaticinteractions between the diffusing molecule and themembrane.88 The pores of the membrane are filledwith a receptor medium so that the transfer of the ac-tive ingredient from the semisolid through the mem-brane involves partitioning into the liquid within thepores (Fig. 2). The kinetics of above-mentioned pro-cess is influenced by the thickness and porosity of the membrane, the viscosity of the receptor, and ad-ditionally the receptor/semisolid partition coefficient.

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 4: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 4/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4035

Table 2.   Selected Membranes used in Release Studies (Co-TranTM, DuraporeTM, 3M, St.Paul, Minnesota)

Membrane Polymer Pore Size (:m) Nature References

Cellulose-based membrane

Benzoylated tubing Regenerated cellulose – Hydrophilic 17,76

Cellulose acetate Cellulose acetate 0.20, 0.45 Hydrophilic 11,32,77,78

Cellulose esters Cellulose esters 0.05, 0.20, 0.45 Hydrophilic 76,79

Cellulose nitrate Cellulose nitrate 0.45 Hydrophilic 76,77,80

Cuprophan Regenerated cellulose 0.45, 0.50 Hydrophilic 17,76,81 Visking Regenerated cellulose – Hydrophilic 17,82–84

Polymeric-based membrane

 AN 69 Polyacrylonitrile – Hydrophilic 17,76

Biodyne Polyamide 0.45 Hydrophilic 76

Supor Polyethersulfone 0.20, 0.45, 0.80 Hydrophilic 35,74,76

Tuffryn Polysulfone 0.45 Hydrophilic 22,23,35,76

Nuclepore Polycarbonate 0.10 Hydrophobic 76

Cyclopore Polycarbonate 0.01, 0.10 Hydrophilic 74,76

Celgard 3500 Polypropylene 0.05 Hydrophobic 76

Folioxane C6 Silicon – Hydrophobic 22

Co-TranTM membrane Microporous polyethylene film – Hydrophobic 45

Nylon Polyamide 0.20, 0.45 Hydrophilic 9,31,32,35

DuraporeTM Polyvinylidene difluoride 0.45 Hydrophilic 10,85

Silicon Polydimethylsiloxane – Hydrophobic 17,86,87

Figure 2.   Mechanism of the active ingredient transfer

from semisolid through the porous membrane into the re-

ceptor fluid.

In order to obtain the best results, the membraneshould have high porosity and minimal thickness.7

In general, cellulose membranes have been widelyused for quality control tests,24,26,89,90 whereas cellu-lose nitrate has been used as a model for the gastricbarrier.91 It is worth mentioning that these mem-branes are reported to be more permeable than bi-ological membranes for allowing the passage of adiffusing molecule regardless of the physicochemicalcharacteristics of the investigated compound.88 Com-

mercially available cellulose membranes contain anumber of softener, preservative, and plasticizer addi-tives, which may have an influence on the diffusion of the active compound. Therefore, the removal of thesesubstances is required. Because they are usually wa-ter soluble, the membrane can be rinsed with dis-tilled water before soaking in the receptor medium.88

However, nonporous membranes such as silicone arealso used in release studies. These membranes arerelatively inert, lipophilic, and provide an ideal en-

 vironment for the permeation of lipophilic molecules.Silicone membranes can be used as a human skinsubstitute when evaluating   in vitro   release char-acteristics of a drug from topical or transdermalformulations.88

In addition, some researchers recommend pretreat-ing the membrane by soaking in the receiving mediumor isopropyl myristate.22,35,45  After adding the sam-ple, the absence of air bubbles underneath the mem-brane should be checked as well.

The diffusion of the active compound from asemisolid dosage formulation into a receiving mediumis determined by the following three processes:

(a) active compound release from the semisoliddosage form itself,

(b) passage of the active compound through themembrane, and

(c) clearance of the active compound from below themembrane.

When neither of the last two processes are ratelimiting, it is only the thermodynamic activity of theinvestigated molecule in the semisolid formulationthat is a significant determinant of drug release.18

If the physical properties of the formulation itself are

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 5: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 5/14

4036   OLEJNIK, GOSCIANSKA, AND NOWAK 

important, diffusion through the base should be therate-limiting step.7 Therefore, it is worth mentioning that the membrane and the receptor medium shouldbe highly permeable and accessible to the active com-pound in the formulation to affect drug release.

Receptor (Receiving Medium)

It is advisable to use a receptor that resembles thephysiological condition of the skin. To maximize par-titioning from the semisolid and transport throughpores of the membrane, the receptor fluid should benonvicious and should exhibit high solvency for theactive ingredient. The pH of medium is also a signifi-cant factor, which should be taken into consideration.The choice of the pH of medium should be based onsuch factors as the pH of the formulation and thepH-solubility profile of the active ingredient and thepH of the target membrane.92 The pH of the recep-tor medium should be adjusted to be in the rangeof 5–6±   0.05 to reflect physiological skin conditions.

 As a rule of thumb, the selected receptor should re-lease a sufficient amount of the active ingredientwithin a reasonable time period.35 The commonlyused receptor medium for water-soluble drugs is anaqueous buffer. However, for products formulatedwith a water-insoluble active compound, the selectionof an appropriate receptor medium to maintain sinkcharacteristics is a challenge.64 In order to facilitateand monitor drug release from such topical formula-tions, it may be necessary to alter the receptor fluid’spH, to add surfactants and/or complexing agents suchas cyclodextrins,16 or to use nonaqueous medium in

which the drug is more soluble.93 The use of hydroal-coholic solutions as the receptor media for lipophilicdrugs such as triamcinoloneacetate,45,94 betametha-sone dipropionate,64 and rooperol tetraacetate11 hasbeen reported.

Sampling Time

In order to determine the release profile of an activecompound, six samples are recommended. Sampling is carried out at regular intervals with the replace-ment of the medium. Sampling points vary depending on the solubility of the active ingredient. However, it

is suggested that samples should be collected over atleast a 6-h period, taking not less than five samples(typical samples intervals are 0.25, 0.5, 1, 2, 4, and6 h), although at times, a duration greater than 24 hmay be required.6

It has been reported that there is a specific timewindow during which samples for release experi-ments should be taken.6 Ideally, samples should beremoved at times when the influence of the membraneand its associated stagnant layer disappears andbefore excessive drug depletion from the semisoliddosage form being tested has taken place. There aretwo reasons to explain the rationale behind withdraw-

ing the samples during this time window. First, it hasbeen shown that the migration of an active compoundfrom a semisolid dosage form to a receptor mediumoccurs as a series of successive diffusional steps.95,96

In the primary step, the membrane, together withan unstirred layer at its surface, has been shown toprovide some resistance to the diffusion of the ac-

tive compound and therefore affects the release rateof the active compound.6 However, in other cases, adiffusional layer can be formed within the semisolidmatrix and can be the rate-controlling element. Con-sequently, sampling time should start when the in-fluence of the membrane and its associated stagnantlayer has disappeared.6 Second, in accordance withtheoretical permeation models, a plot of the amountof active compound released versus the square rootof time should be linear if drug release from a for-mulation is diffusion controlled or if diffusion is therate-controlling element within the dosage form.95,96

It has been reported that active compound releasefrom semisolid dosage forms may deviate from lin-earity at extended times, and the deviation usuallyoccurs when more than approximately 35%–45% of the investigated molecule has been released.6

Temperature of Analysis

In case of semisolid dosage forms, the test temper-ature is typically set at 32±   0.5◦C to reflect theusual skin temperature. Deviations might be justi-fied when products are for specific sites of action;for example, vaginal creams may be tested at 37 ±0.5◦C.11,16,35,90,97 However, the use of excessively high

temperatures may melt the base or the product being tested or cause significant physical changes to sucha product, which in turn may change the resistanceof the matrix to active compound diffusion into thereceptor medium and should, therefore, be avoided.6

Chattaraj and Kanfer32 investigated the effect of the temperature on the release characteristics of acy-clovir from different creams.32 The release of acy-clovir, using a cellulose acetate as the membrane,was measured at 32◦C and 37◦C. The results indicatethat this relatively small difference in temperatureappeared to have a significant effect on the release

of the active compound from creams. The tempera-ture effect is probably because of the nature of thesemisolid dosage forms, which would account for thedifference in release seen as a result of the heat sensi-tivity of the components of the creams. Vehicles usedto formulate semisolid dosage forms would becomeless viscous with increasing temperature and therebyfacilitate the release of the drug.32

APPARATUS FOR IVRT

The choice of the apparatus used in release studies de-pends on the physicochemical properties of the dosage

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 6: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 6/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4037

formulations. All parts of the devices that may con-tact with the formulation or medium should be chem-ically inert and, therefore, should not react with thetest product. Moreover, all metal parts of the devicesmust be made from a suitable stainless steel or coatedwith an appropriate material.98

Franz Cell and its Modifications

The Franz cell apparatus (known as the verticaldiffusion cell) has been the standard system usedfor the study of semisolid drug formulations since19785,11,16,24,29,64,90,94,99–101 and is recommended bythe USP.102 This device consists of two primary cham-bers—a jacket glass receptor (typically 12.5mL in vol-ume) and a glass sampling port. These chambers areseparated by a membrane that is usually synthetic,although an animal skin can be used. A schematicpicture of the Franz diffusion cell is shown inFigure 3. The test product should be added in a con-trolled amount to the upper side of the membrane inthe top chamber.6 The bottom chamber is filled withthe sampling fluid from which the samples are col-lected at regular intervals for the analysis. The exper-iment determines the amount of the active ingredientthat has been released through the membrane at eachtime point. The main advantages of the Franz cell areits ease of operation and its large sample size, whichensures consistent results. Moreover, the technique issimple, reliable, reproducible, and suitable for in situ

gels, creams, and ointments measurements. One hasto keep in mind that air bubbles at the membrane–liq-uid interface cause poor precision in the data. The

choice of the right membrane is crucial because themembrane can also control the diffusion of the activecompound.

 According to Keshary and coworkers,103,104 the de-sign of the Franz cell does not provide for adequate so-lution hydrodynamics, appropriate mixing efficiency,and the temperature control that are necessary for

Figure 3.   Schematic picture of the typical Franz diffusion

cell.78

quantitative permeation tests. Therefore, they intro-duced several modifications to the original Franz cellapparatus. In comparison with the receptor compart-ment of the original Franz cell, their newly devel-oped diffusion cell has a shorter cylindrical receptorcompartment and is completely enclosed by a water

 jacket. A star-headed magnetic stirrer is employed to

agitate the receptor medium. Because of these modi-fications, the equilibrium temperature of the receptormedium is better maintained. Moreover, the solution-mixing efficiency is improved, and the thickness of the boundary diffusion layer is decreased.103

The Hanson Microette (Hanson Research Corpo-ration, Chatsworth, CA) employs six vertical, au-tomated Franz diffusion cells that have 15mmdiameters.6 Figure 4 presents sampling from one cell.The Hanson Microette system simultaneously sam-ples and collects from all six cells. Each cell is filledwith a receptor solution from the medium beaker. Theamount of test formulation is checked and then placedon the membrane in the cell. On program command,the stirrer stops working, and the appropriate amountof the aliquot of fresh replacement medium is injectedfrom the precision syringe pump into the capillarymedium. The sample lines are cleaned by a wash cy-cle just before the sampling takes place.6

Paddle-Over-Disk

 Another technique that can be used is the USP No.5 [European Pharmacopeia (PhEur) 2.9.4.1] paddle-over-disk method.5 The formulation is filled into theUSP stainless steel disk and placed on the bottom of a

release vessel filled with the receptor medium (Fig. 5).The analysis is performed by using stirring paddles(25 mm above the surface of the disk) at 50 rpm. Inthis method, the membrane is not required. Only thestainless steel is employed to perform the role of thebarrier. The main disadvantage of this technique isthe possibility for the formulation to dissolve in the re-ceptor medium. Thus, dissolution is measured ratherthan release. However, this method could be usefulto carry out release testing of products that do notdissolve in the receptor medium.105

Enhancer Cell Assembly

Enhancer cell, designed by Vankel Technology Group(Cary, North Carolina), is another apparatus that isused for release testing of semisolid formulations.106

The enhancer method resembles the paddle-over-disktechnique. However, in this case, the use of a mem-brane is required. In addition, the paddle over extrac-tion cell method is recommended by PhEur 2.9.4.2.The Vankel release system comprises a bath with six

 vessels (200 mL). A circulator pump can be used toattain a constant temperature. The inner diameter of the Vankel enhancer cell is 21 mm, which gives a sur-face area of 345.44mm2.78  An enhancer cell composed

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 7: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 7/14

4038   OLEJNIK, GOSCIANSKA, AND NOWAK 

Figure 4.   Schematic representation of Hanson Microette sampling system (redrawn from

Ref. 6).

Figure 5.   Apparatus No. 5–-paddle-over-disk. The disk

assembly is used to hold the system flat and is positioned

parallel with the bottom of the paddle blade.102

of Teflon consists of a cap, an O-ring, a membrane, anda drug reservoir (Fig. 6). The dosage volume of thecell body is adjusted with a special tool that is used

to increase or decrease the depth of the reservoir. Thecell is filled with the product to be tested, and thenthe membrane and O-ring are placed over the sam-ple surface. The top cap is screwed on, and a specialalignment tool is used to position the O-ring and to fitthe membrane securely to the sample. The assemblyis available with 4.0, 2.0, or 0.5 cm2 surface areas. Theadvantage of the enhancer cell method is its availabil-ity to most researchers and manufacturers45 becausea basic USP dissolution apparatus is used. The large

 volume range makes it easier to adapt this systemto study the release of products containing low con-centrations of active ingredients or ingredients thatare difficult to analyze.90 Moreover, the method re-quires fewer accessories and reduces the time andcost needed for setting up the equipment. The appli-cation of enhancer cells made of Teflon prevents theformulation from interacting with the cell. Such prob-lems can occur when using glass diffusion cells.45 Thedisadvantage of the enhancer cell is that the temper-ature equilibrium between the formulation and thereceptor phase could take a finite time because Teflon

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 8: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 8/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4039

Figure 6.   Schematic picture of the enhancer cell with its

elements (on the left). On the right, the enhancer cell inside

the vessel.6

Figure 7.   Picture of inverted rotating cylinder.105

is a poor conductor of heat with a small heat transfer

coefficient. This requires the cells and the formula-tion to be stored at the study temperature before use.This temperature control is especially important dur-ing the evaluation of drug release from ointments thattake a longer time to equilibrate.45

Inverted Enhancer Cell

In this method, a machined, round cavity of Teflon orstainless steel is used as the holder of the test prod-uct (Fig. 7). The cylinder filled with formulations isplaced into a standard release vessel and rotated, usu-ally at 50 rpm.105 In contrast to the above-mentionedmethods, the use of a membrane is not required. Fur-thermore, this method can be used only with productsthat do not dissolve in the receiving medium. It is es-pecially useful in the case of hydrophobic ointmentbases comprising the solubilized drug.

The Rotating Cylinder Method (USP Apparatus 6/PhEur2.9.4.3)

This method is similar to the USP basket-type disso-lution apparatus, except that the system is attachedto the surface of a hollow cylinder immersed in amedium at 32±   0.5◦C.5,104 The shaft and cylindercomponents of the stirring element are fabricated

from stainless steel to the specifications shown inFigure 8. The dosage unit is placed on the cylinderat the beginning of each test. The distance betweenthe inside bottom of the vessel and the cylinder ismaintained at 25± 2 mm during the test.5,102

The Flow-Through Cell Diffusion Apparatus

In flow-through cells, the receptor medium con-tinuously flows through the acceptor compart-ment, and, as a result, the medium is refreshedcontinually.89,90,107 The apparatus consists of a reser-

 voir with a release medium, a pump, and a waterbath (Fig. 9). The pump is used to force the releasemedium upward through the vertically placed flow-through cell. The bottom part is filled with glass beads(1 mm diameter) and with one additional bead (5 mmdiameter), which is employed to protect the fluid en-try tube. In order to measure the release rate of theactive compound from a semisolid formulation, an in-

sertion cell is placed inside the flow-through cell. Thedissolution liquid is generally collected in a separatereservoir as it leaves the insertion cell. Fractions areremoved at specified intervals and analyzed. The flow-through cell device is specifically used for lipophilicsolid dosage forms.89,90,107

The application of an “insertion cell” offers distinctadvantages compared with the Franz cells because itis easier to use and readily adaptable for use with theflow-through apparatus. Moreover, it does not sufferfrom the problem of having to remove air bubbles atthe membrane–liquid interface, which commonly oc-

curs when using Franz cells.

89

 Additionally, it should be mentioned that typicalIVRT apparatus consist of six cells. For each run, thetwo products (reference product and test product) be-ing compared should be placed in six cells accord-ing to the pattern illustrated in the Figure 10. Thisprocedure will help to ensure an unbiased compari-son in the event of a systematic error difference be-tween runs. The assignment of products to cells (i.e.,whether the reference product or the test product isplaced in the “upper left corner cell” of the appara-tus) can be made randomly or systematically (i.e.,alternating the pattern for each successive run). In

the case of nonstandard equipment, which consists of other than six cells, the same pattern of experimentalruns is still valid. It should be mentioned that in ap-paratus with only a single cell, the runs of referencetests and product tests should be intermixed.108

KINETICS OF IVRT

The release from delivery systems can be followed bythree main models, that is, Higuchian kinetics, zero-order kinetics, and first-order kinetics, taking into ac-count the Fickian diffusion laws.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 9: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 9/14

4040   OLEJNIK, GOSCIANSKA, AND NOWAK 

Figure 8.   Cylinder stirring element. All measurements are expressed in cm unless noted

otherwise (redrawn from Ref. 102).

Figure 9.   Schematic picture of flow-through cell for semisolids forms (on the left). On the

right, schematic diagram of a flow-through dissolution apparatus.78

Figure 10.   Experimental run according to SUPAC-SS guidance (different cell assignment a

and b). Test represents the postchange lot (Test product) and reference represents the prechange

lot (reference product).108

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 10: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 10/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4041

Modeling Diffusion

Diffusion can occur in a double diffusion cell, wherea concentrated solute solution is separated from thereceptor fluid by a semipermeable membrane.109,110

During the diffusion process, the solute passes from aregion of high concentration (donor compartment) toa region of low concentration (receptor compartment)through a membrane as a barrier. The decrease of thesolute concentration in the donor compartment is fol-lowed by the increase in the concentration in the ac-ceptor compartment. Until an equilibrium is reached,the process proceeds and a steady state is reached.111

The diffusion coefficient through the semipermeablemembrane can be measured by kinetic methods whenthe concentrations in the donor and acceptor com-partments are checked.112 Diffusion coefficients canbe defined by using Fick’s first and second laws of dif-fusion, which are appropriate when a membrane actsas a rate-controlling step for the release of bioactive

molecules.

Fick’s First Law 

The diffusion of molecules is define as rate of masstransfer (d M  /dt) and is expressed as flux ( J ). Flux isdefined as the rate of mass transfer through a unitsurface area of a barrier (membrane).113,114

 J = 0.5d M 

 Sdt

where d M —change in mass, g, S–barrier surface area,

cm2

, dt–change in time, s. Flux units g s-1

cm-2

and areusually expressed as kg m−2 s−1.There is a direct correlation between the flux and

the concentration gradient. This relationship is ex-pressed by the following equation:

 J = − DdC

d x

where dC–change in concentration of the solute, D—diffusion coefficient of the solute cm2 /s, and d x-–change in distance, cm.

Fick’s Second Law 

In case of Fick’s second law, the change in concen-tration with time in a definite spatial region ( x, y, z) istaken into account. This law states that the change inconcentration with time at a specific location is pro-portional to change in the concentration gradient atthat point in time.

∂C

∂t  =  D

∂2C

∂ x2  +

∂2C

∂ y2  +

∂2C

∂ z2

where x, y, and z  are the spatial coordinates.

Table 3.   Equations for Release Kinetics111

Model Equation

Zero order   F = kt

First order ln F = kt

Higuchian   F = kt1/2

 F —the fraction of active compound released from semisolid,  k-–therelease constant, and t—time.

Higuchian Model

The amount of the released active compound changeswith the appropriate rate during the analysis. The re-lease of water-soluble and poorly water-soluble drugsfrom semisolids was studied by Higuchi.18,96 Today,the Higuchi equation is considered to be one of thewidely used and the most well-known controlled-release equations. According to him, the release of a drug depends upon its diffusion.

His general equation is given below:

 M t

 M 0=  K 

1/2t

where   M t—amount of drug released in time   t, M 0—initial amount of the drug released, and t—timerequired.

In addition, two theoretical models have been sug-gested, one in which the active compound exists asa suspension in the base (Model I) and the other inwhich the active compound exists as a solution in thebase (Model II). In accordance with Higuchi,1896,115

the resulting plot should be linear for the controlled

release of the active component in the suspension:

Q  = (2C0Cs Dt)1/2,   Cs  << C0

or in solution

Q  = 2C0

 Dt

B

1/2

where  Q–amount of the drug released per unit areaof sample, C0–initial concentration of the drug in thesemisolid,  Cs–solubility of the drug in the semisolid

matrix,   D–diffusion coefficient of the drug in thesemisolid, and   t–expired time. A plot of the amountof the active compound released per unit membranearea (mg/cm2) versus the square root of time shouldresult in a straight line. The slope of the line presentsthe release rate of the active component. The plot ob-tained from the kinetic data shows that the efficiencyof the active compound released from the semisoliddosage form (Table 3) .

The zero-order release model is similar to theHiguchian kinetics; however, the release remains con-stant over a period of time because the releasing sur-face is constant. The zero-order model refers to the

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 11: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 11/14

4042   OLEJNIK, GOSCIANSKA, AND NOWAK 

assumption that the average pore size in the deliverysystem is smaller than the size of the investigatedcompound. In practice, zero-order release is usuallyconverted to the first-order model.116,117

CONCLUSIONS

The controlled release of active compounds fromsemisolid dosage forms is a significant factor in esti-mating the efficiency of the active compounds. There-fore, controlled-release studies have been carried outin quality control laboratories. Such research permitsus to assess a product’s activity after introducing anychanges in composition, in apparatus, or in the pro-duction process. Release tests of drugs from creams,gels, and ointments, in conjunction with solubility,particle size, and crystalline form, are important fora proper characterization of newly developed prod-ucts, and thus their potential therapeutic effect can

be predicted. For this reason, it is essential to se-lect the appropriate release test conditions mentionedin this article (temperature, sampling time, receptor,membrane, and type of apparatus).

ACKNOWLEDGMENTS

The authors would like to thank the National Sci-ence Center for the financial support (N N204 403040;2011–12).

REFERENCES

1. Sathe PM, Raw AS, Ouderkirk LA, Yu LX, Husain AS. 2005.

Drug product performance,   in vitro. In Generic drug prod-

uct development: Solid oral dosage forms; Shargel L, Kan-

fer I, Eds. New York City, New York: Marcel Dekker, Inc.,

pp. 187–209.

2. Azarmi S, Roa W, Lobenberg R. 2007. Current perspectives

in dissolution testing of conventional and novel dosage forms.

Int J Pharm 328:12–21.

3. Hanson WA. 1982. Overall considerations. In Handbook of 

dissolution testing; Hanson WA, Ed. Corvallis, Oregon: Phar-

maceutical Technology Publications, pp 1–11.

4. FDA SUPAC SS working group. 1997. Guidance for indus-

try. Nonsterile semisolid dosage forms. Scale-up and postap-

proval changes: Chemistry, manufacturing and controls;

in vitro  release testing and in vivo  bioequivalence documen-tation..

5. Ueda CT, Shah VP, Derdzinski K, Ewing G, Flynn G,

Maibach H, Marques M, Rytting H, Shaw S, Thakker K,

 Yacobi A. 2009. Topical and transdermal drug products.

Pharmacopeial Forum 35:750–764.

6. Zatz JL, Segers JD. 1998. Techniques for measuring in vitro

release from semisolids. Dissolution Technol 5:3–13.

7. Zatz JL. 1995. Drug release from semisolids: Effect of mem-

brane permeability on sensitivity to product parameters.

Pharm Res 12:787–789.

8. Bonacucina G, Cespi M, Misici-Falzi M, Palmieri GF.

2006. Rheological, adhesive and release characterisation

of semisolid Carbopol/tetraglycol systems. Int J Pharm

307:129–140.

9. Yang Y, Wang S, Xu H, Sun Ch, Li X, Zheng J. 2008. Prop-

erties of topically applied organogels: Rheology and  in vitro

drug release. Asian J Pharm Sci 3:175–183.

10. Sawant PD, Luu D, Ye R, Buchta R. 2010. Drug release

from hydroethanolic gels. Effect of drug’s lipophilicity (log 

 P), polymer–drug interactions and solvent lipophilicity. Int J

Pharm 396:45–52.

11. Csoka I, Csanyi E, Zapantis G, Nagy E, Feher-Kiss A,

Horvath G. 2005.   In vitro   and   in vivo   percutaneous ab-sorption of topical dosage forms: Case studies. Int J Pharm

291:11–19.

12. Buhse L, Kolinski R, Westenberger B, Wokovich A, Spencer

J, Chen C, Turujman S, Gautam-Basak M, Kang G, Kibbe A,

Heintzelman B, Wolfgang E. 2005. Topical drug classifica-

tion. Int J Pharm 295:101–112.

13. Bhavsar JD, Brhambhatt VG, Patel MR, Patel KR, Patel NM.

2011. Novel approaches in semisolids. IntJ PharmWorld Res

2:1–22.

14. Gupta P, Garg S. 2002. Recent advances in semisolid

dosage forms for dermatological application. Pharm Technol

26:144–162.

15. Morell JL, Claramonte MD, Vialard A. 1996. Validation of 

a release diffusion cell for topical dosage forms. Int J Pharm

137:49–55.16. Corbo M, Shultz TW, Wong GK, Van Buskirk GA. 1993. De-

 velopment and validation of in vitro release testing meth-

ods for semisolid formulations. Pharm Technol 17:112–

128.

17. Ng SF, Rouse JJ, Sanderson FD, Meidan V, Eccleston

GM. 2010. Validation of a static Franz diffusion cell sys-

tem for in vitro permeation studies. AAPS PharmSciTech

11:1432–1441.

18. Higuchi T. 1961. Rate of release of medicaments from oint-

ment bases containing drugs in suspension. J Pharm Sci

50:874–875.

19. Roseman TJ, Higuchi WI. 1970. Release of medroxypro-

gesterone acetate from a silicone polymer. J Pharm Sci

59:354–357.

20. Tojo K. 1985. Intrinsic release rate from matrix-type deliverysystems. J Pharm Sci 74:685–687.

21. Bottari F, Di Colo G, Nannipieri E, Saettone MF, Serafini MF.

1977. Release of drugs from ointment bases II: Benzocaine

from suspension-type gels. J Pharm Sci 66:926–931.

22. Clement P, Laugel C, Marty JP. 2000. Influence of three

synthetic membranes on the release of caffeine from concen-

trated W/O emulsions. J Control Release 66:243–254.

23. Clement P, Laugel C, Marty JP. 2000. In vitro release of caf-

feine from concentrated W/O emulsions: Effect of formulation

parameters. Int J Pharm 207:7–20.

24. Dias M, Farinfa A, Faustino E, Hadgraft J, Pais J, Toscano

C. 1999. Topical delivery of caffeine from some commercial

formulations. Int J Pharm 182:41–47.

25. Bolzinger MA, Briancon S, Pelletier J, Fessi H, Chevalier

 Y. 2008. Percutaneous release of caffeine from microemul-sion, emulsion and gel dosage forms. Eur J Pharm Biopharm

68:446–451.

26. Shah VP, Elkins J, Hanus J, Noorizadeh C, Skelly JP. 1991.

In vitro release of hydrocortisone from topical preparations

and automated procedure. Pharm Res 8:55–59.

27. Shah VP, Elkins J, Skelly JP. 1989. Determination of in

 vitro drug release from hydrocortisone creams. Int J Pharm

53:53–59.

28. Niemi L, Kahela P, Turakka L. 1989. Effect of water content

and type of emulgator on the release of hydrocortisone from

o/w creams. Acta Pharm Nord 1:23–30.

29. Sanghvi PP, Collins CC. 1993. Comparison of diffusion stud-

iesof hydrocortisonebetweenthe Franz cell andthe enhancer

cell. Drug Dev Ind Pharm 19:1573–1585.

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 12: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 12/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4043

30. Laugel C, Baillet A, Piemi MPY, Marty JP, Ferrier D. 1998.

Oil–water–oil multiple emulsions for prolonged delivery of 

hydrocortisone after topical application: Comparison with

simple emulsions. Int J Pharm 160:109–117.

31. Christensen JM, Chuong MC, Le H, Pham L, Bendas E.

2011. Hydrocortisone diffusion through synthetic membrane,

mouseskin, and EpidermTM cultured skin. Arch Drug Inform

4:10–21.

32. Chattaraj SC, Kanfer I. 1995. Release of acyclovir from semi-solid dosage forms: A semi-automated procedure using a sim-

ple plexiglass flow-through cell. Int J Pharm 125:215–222.

33. Hwang YJ, Oh C, Oh SG. 2005. Controlled release of retinol

from silica particles prepared in O/W/O emulsion—The effect

of surfactants and polymers. J Control Release 106:339–349.

34. Kundu SC, Cameron AD, Meltzer NM, Quick TW. 1993.

Development and validation of method for determination of 

in vitro release of retinoic acid from creams. Drug Dev Ind

Pharm 19:425–438.

35. Thakker KD, Chern WH. 2003. Development and validation

of in vitro release tests for semisolid dosage forms—Case

study. Dissolution Technol 10 10–15.

36. Gueroll Z, Hekimoglu S, Demirdamar R, Sumnu M. 1996.

Percutaneous absorption of ketoprofen. I. In vitro release and

percutaneous absorption of ketoprofen from different oint-ment bases. Pharm Acta Helv 71:205–212.

37. Chi SC, Jun HW. 1991. Release rates of ketoprofen from

poloxamer gels in a membraneless diffusion cell. J Pharm

Sci 80:280–283.

38. Gallagher SJ, Trottet L, Heard CM. 2003. Ketoprofen: Re-

lease from, permeation across and rheology of simple gel

formulations that simulate increasing dryness. Int J Pharm

268:37–45.

39. Tas C, Ozkan Y, Savaser A, Baykara T. 2003. In vitro release

studies of chlorpheniramine maleate from gels prepared by

different cellulose derivatives. Il Farmaco 58:605–611.

40. Velissaratou AS, Papaioannou G. 1989. In vitro release of 

chlorpheniramine maleate from ointment bases. Int J Pharm

52:83–86.

41. Babar A, Bhandari RD, Plakogiannis PM. 1991. In-vitro re-leasestudies op chlorpheniramine maleate from topical bases

using cellulose membrane and hairless mouse skin. DrugDev

Ind Pharm 17:1027–1040.

42. Bajpai SK, Dubey S. 2005. In vitro dissolution studies for

release of vitamin B12   from poly( N -vinyl-2-pyrrolidone-co-

acrylic acid) hydrogels. React Funct Polym 62:93–104.

43. Bottari F, DeColo G, Nannipieri E, Saettone MF, Serafini

MF. 1974. Influence of drug concentration on   in vitro   re-

lease of salicylic acid from ointment bases. J Pharm Sci

63:1779–1783.

44. Chowhan ZT, Pritchard R. 1975. Release of corticoids from

oleaginous bases containing drug in suspension. J Pharm Sci

64:754–759.

45. Rege PR, Vilivalam VD, Collins CC. 1998. Development

in release testing of topical dosage forms: Use of the En-hancer CellTM with automated sampling. J Pharm Biomed

 Anal 17:1225–1233.

46. Segers JD, Zatz JL, Shah VP. 1997. In vitro release of phenol

from ointment formulation. Pharm Technol 21:70–81.

47. Badkar A, Talluri K, Tenjarla S, Jaynes J, Banga AK. 2000.

In vitro release testing of a peptide gel. Pharm Technol

24:44–52.

48. Rahman MS, Babar A, Patel MK, Plakogiannis FM. 1990.

Medicament release from ointment bases: V. Naproxen in-

 vitro release and in-vivo percutaneous absorption in rabbits.

Drug Dev Ind Pharm 16:651–672.

49. Attia DA. 2009. In vitro and in vivo evaluation of transder-

mal absorption of naproxen sodium. Aust J Basic Appl Sci

3:2154–2165.

50. FerreiraRAM, Seiller M, GrossiordJL, Marty JP,Wepierre J.

1994. Vehicle influence on in vitro release of metronidazole:

Role of w/o/w multiple emulsion. Int J Pharm 109:251–259.

51. Muktadir A, Babar A, Cutiea AJ, Plakogiannis FM. 1986.

Medicament release from ointment bases: III. Ibuprofen: In-

 vitro release and in-vivo absorption in rabbits. Drug Dev Ind

Pharm 12:2521–2540.

52. Rasool BKA, Abu-Gharbieh EF, Fahmy SA, Saad HS, Khan

SA. 2010. Development and evaluation of ibuprofen trans-dermal gel formulations. Top J Pharm Res 9:355–363.

53. GopiC, Kumar TMV, Dhanaraju MD. 2010. Designand eval-

uation of novel ibuprofen gel and its permeability studies. Int

J Biopharm 1:82–84.

54. Fang JY, Hwang TL, Leu YL. 2003. Effect of enhancers and

retarders on percutaneous absorption of flurbiprofen from

hydrogels. Int J Pharm 250:313–325.

55. El Gendy AM, Jun HW, Kassem AA. 2002. In vitro release

studies of flurbiprofen from different topical formulations.

Drug Dev Ind Pharm 28:823–831.

56. Pandey MS, Belgamwar VS, Surana SJ. 2009. Topical deliv-

ery of flurbiprofen from Pluronic lecithin organogel. Indian J

Pharm Sci 71:87–90.

57. Bonina FP, Montenegro L. 1994.Vehicle effects on in-vitro

heparin release and skin penetration from different gels. IntJ Pharm 102:19–24.

58. Gutowska A, Bae YH, Feijen J, Kim SW. 1992. Heparin

release from thermosensitive hydrogels. J Control Release

22:95–104.

59. Hinrichs WJL, ten Hoopen HWM, Wissink MJB, Engbers

GHM, Feijen J. 1997. Design of a new type of coating for the

controlled release of heparin. J Control Release 45:163–176.

60. Ahola MS, Sailynoja ES, Raitavuo MH, Vaahtio MM, Salonen

JI,Yli-UrpoAUO. 2001. In vitro release of heparin from silica

xerogels. Biomaterials 22:2163–2170.

61. Barry BW. 1983. Dermatological formulations. Percutaneous

absorption. In Drugs and the pharmaceutical sciences; Swar-

brik, Ed. Vol. 18. New York City, New York: Marcel Dekker,

Inc., pp 49–94.

62. Flynn GL, Shah VP, Tenjarla SN, Corbo M, DeMagistris D,Feldman TG. 1999. Assessment of value and applications

of in vitro testing of topical dermatological drug products.

Pharm Res 16:1325–1329.

63. Shah VP, Elkins J, Shaw S, HansonR. 2003. In vitro release:

Comparative evaluation of vertical diffusion cell system and

automated procedure. Pharm Dev Technol 8:97–102.

64. Shah VP, Elkins J, Williams RL. 1999. Evaluation of the test

system used for  in vitro  release of drugs for topical dermato-

logical drug products. Pharm Dev Technol 4:377–385.

65. Shah VP, Elkins J. 1995. In vitro release from corticosteroid

ointments. J Pharm Sci 84:1139–1140.

66. Baker RW, Heller J. 1989. Material selection for transder-

mal delivery systems. In Transdermal drug delivery: Devel-

opment issues and research initiatives; Hadgraft J, Guys

RH, Eds. New York City, New York: Marcel Dekker, Inc.,pp 293–311.

67. Guyot M, Fawaz F. 2000. Design and in vitro evaluation of 

adhesive matrix for transdermal delivery of propranolol. Int

J Pharm 204:171–182.

68. Gabiga H, Cal K, Janicki S. 2000. Effect of penetration en-

hancers on isosorbide dinitrate penetration through rat skin

from a transdermal therapeutic system. Int J Pharm 199:1–6.

69. Minghetti P, Cilurzo F, Casiragh A, Molla FA, Montanari L.

1999. Dermal patches for controlled release of miconazole: In-

fluence of drug concentration on the technical characteristics.

Drug Dev Ind Pharm 25:679–684.

70. Tsai CJ, Hu LR, Fang JY, Lin HH. 1999. Chitosan hydro-

gel as a base for transdermal delivery of berberine and its

evaluation in rat skin. Biol Pharm Bull 22:397–401.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012

Page 13: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 13/14

4044   OLEJNIK, GOSCIANSKA, AND NOWAK 

71. Gale R, Spitze LA. 1981. Permeability of camphor in ethy-

lene vinyl acetate copolymers. In Proceedings of the Eighth

International Symposium on Controlled Release of Bioactive

Materials. Minneapolis, Minnesota: Controlled Release Soci-

ety, p 183.

72. Boretos JW, Detmer DE, Donachy JH. 1971. Segmented

polyurethane: A polyether polymer II. Two year experience.

J Biomed Mater Res 5:373–387.

73. Brain KR, Walters KA, Green DM, Brain S, Loretz LJ,Dressler WE. 2005. Percutaneous penetration of di-

ethanolamine through human skin in vitro: Application from

cosmetic vehicles. Food Chem Toxicol 43:681–690.

74. Gabbanini S, Matera R, Beltramini C, Minghetti A,

 Valgimigli L. 2010. Analysis of in vitro release through re-

constructed human epidermis and synthetic membranes of 

multi-vitamins from cosmetic formulations. J Pharm Biomed

 Anal 52:461–467.

75. Branka R, Mirjana G, Estelle T-T, Fabrice P, Francoise G.

2009. Simultaneous absorption of vitamins C and E from top-

ical microemulsions using reconstructed human epidermis as

a skin model. Eur J Pharm Biopharm 72:69–75.

76. Ng SF, Rouse J, Sanderson D, Eccleston G. 2010. A com-

parative study of transmembrane diffusion and permeation

of ibuprofen across synthetic membranes using Franz Diffu-sion Cells. Pharmaceutics 2:209–223.

77.  Ozer O,KıvcakB, Mutlu B,Akay S,Sa lam H,Tomek S. 2007.

In vitro release studies on multiple and simple emulsions of 

tocopherol with Pistacia leaves. Sci Pharm 75:97–109.

78. Rapedius M, Blanchard J. 2001. Comparison of the

Hanson Microettet and the Van Kel Apparatus for  In Vitro

Release Testing of Topical Semisolid Formulations. Pharm

Res 18:1440–1447.

79. Edgar KJ, Buchanan CM, Debenham JS, Rundquist PA,

Seiler BD, Shelton MC, Tindall D. 2001. Advances in cel-

lulose ester performance and application. Prog Polym Sci

26:1605–1688.

80. Arellano A, Santoyo S, Martn C, Ygartua P. 1998. Surfactant

effects on the in vitro percutaneous absorption of diclofenac

sodium. Eur J Drug Metab Pharmacokinet 2:307–312.81. Djordjevic L, Primorac M, Stupar M. 2005. In vitro  release

of diclofenac diethylamine from caprylocaproyl macrogolglyc-

erides based microemulsions. Int J Pharm 296:73–79.

82. Conaghey OM, Corish J, Corrigan OI. 1998. The release of 

nicotine from a hydrogel containing ion exchange resins. Int

J Pharm 170:215–224.

83.  Ozsoy Y,Gung or S, Cevher E. 2004.Vehicle effects on in vitro

release of tiaprofenic acid from different topical formulations.

Farmaco 59:563–566.

84. Smith EW, Haigh J. 1992. In vitro diffusion cell and valida-

tion. Acta Pharm Nord 4:171–178.

85. Yoshida H, Tamura S, Toyoda T, Kado K, Ohnishi N, Ibuki

R. 2004. In vitro release of Tacrolimus from Tacrolimus oint-

ment and its speculated mechanism. Int J Pharm 270:55–64.

86. Khan G, Frum Y, Sarheed O, Eccleston GM, Meidan VM.2005. Assessment of drug permeability distribution in two

different model skins. Int J Pharm 303:81–7.

87. Frum Y, Eccleston GM, Meidan VM. 2007. Evidence that

drug flux through synthetic membranes is assocated with

normally distributed permeability coefficients. Eur J Pharm

Biopharm 67:434–9.

88. Haigh JM, Smith EW. 1994. The selection and use of natural

and synthetic membranes for  in vitro diffusion experiments.

Eur J Pharm Sci 2:311–330.

89. Chattaraj SC,Kanfer I. 1996. The “insertion cell”:A novelap-

proach to monitor drug release from semi-solid dosage forms.

Int J Pharm 133:59–63.

90. Liebenberg W, Engelbrecht E, Wessels A, Evarakonda B,

 Yang W, De Villie MM. 2004. A comparative study of the

release of active ingredients from semisolid cosmeceuticals

measured with Franz, enhancer or flow-through cell diffu-

sion apparatus. J Food Drug Anal 12:19–28.

91. Ceschel GC, De Filippis P. 1981. Diffusion through artificial

lipid barriers of two forms of betamethasone and correlation

with in vivo data. Pharm Acta Helv 56:291–295.

92. Van Amerongen IA, De Ronde HAG, Klooster NTM.

1992. Physical–chemical characterization of semisolid topi-

cal dosage form using a new dissolution system. Int J Pharm86:9–15.

93. Skelly JP, Shah VP, Maibach HI, Guy RH, Wester RC, Flynn

G. 1987. FDAand AAPS report of theworkshop on principles

and practices of in vitro percutaneous penetration studies:

Relevance to bioavailability and bioequivalence. Pharm Res

4:265–267.

94. Fares MH, Zatz LJ. 1995. Measurement of drug release from

topical gels using two types of apparatus. Pharm Technol

19:53–58.

95. Higuchi T. 1963. Mechanism of sustained-action medication.

Theoretical analysis of rate of release of solid drugs dispersed

in solid matrices. J Pharm Sci 52:1145–1149.

96. Higuchi WI. 1962. Analysis of data on the medicament re-

lease from ointments. J Pharm Sci 51:802–804.

97. Siewert M, Dressman J, Brown CK, Shah VP. 2003. FIP/  AAPS guidelines to dissolution/in vitro release testing of 

novel/special dosage forms. AAPS PharmSciTech 4:1–10.

98. European Pharmacopoeia 5.0, 2.9.3. 2005. Dissolution test

for solid dosage forms.

99. Franz TJ. 1978. The finite dose technique as a valid in vivo

model for the study of percutaneous absorption. Curr Probl

Dermatol 7:58–68.

100. Pillai R, Shah VP,Albrecht CF,Chattaraj SC,FlynnG. 2001.

Release of hydrocortisone from a cream matrix: Dependency

of release on suspension concentration and measurement of 

solubility and diffusivity. Pharm Dev Technol 6:373–384.

101. Vlachou MD, Rekkas DM, Dallas PP, Choulis NH. 1992. De-

 velopment and in vitro evaluation of griseofulvin gels using 

Franz diffusion cells. Int J Pharm 82:47–52.

102. Ueda CT, Shah VP, Derdzinski K, Ewing G, Flynn G,Maibach H, Marques M, Rytting H, Shaw S, Thakker K,

 Yacobi A. 2010. Topical and transdermal drug products. Dis-

solution Technol 17 12–25.

103. Keshary PR, Chien YW. 1984. Mechanisms of transdermal

controlled nitroglycerinadministration (I): Development of a

finite-dosing skin permeation system. Drug Dev Ind Pharm

10:883–913.

104. Flynn LG, Keshary PR, Huang YC. 1983. Comparative con-

trolled skin permeation of nitroglycerin frommarketedtrans-

dermal delivery systems. J Pharm Sci 72:968–970.

105. Corbo M. 1995. Techniques for conducting in vitro release

studies on semisolid formulations. Dissolution Technol 2:3–6.

106. Vankel Technology Group Buyer’s Guide. 2005. p 37.

107. Tanjo H, Roemele PEH, Van Veen GH, Stieltjes H, Junginger

H, Bodde HE. 1997. Newdesign of a flow-through permeationcell for studying in vitro permeation studies across biological

membrane. J Control Release 45:41–47.

108. Marangon A, Bock U, Haltner E. 2009. In vitro release test-

ing for semisolid formulations. Across Barriers 1–5.

109. Liang S, Zhang L, Xu J. 2007. Morphology and permeability

of cellulose/chitin blend membranes. J Membr Sci 287:19–28.

110. Tenjarla SN, Kasina R, Puranajoti P, Omar MS, Harris

WT. 1999. Synthesis and evaluation of N-acetylprolinate

esters—Novel skin penetration enhancers. Int J Pharm

192:147–158.

111. Kasapis S, Norton IT, Ubbik JB. 2009. Modern biopoly-

mer science. Bridging the divide between fundamental trea-

tise and industrial application (chapter 16). Amsterdam, The

Netherlands: Elsevier, pp 519–557.

 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012 DOI 10.1002/jps

Page 14: Active Compounds Release From Semisolid Dosage Forms

7/21/2019 Active Compounds Release From Semisolid Dosage Forms

http://slidepdf.com/reader/full/active-compounds-release-from-semisolid-dosage-forms 14/14

 ACTIVE COMPOUNDS RELEASE FROM SEMISOLID DOSAGE FORMS   4045

112. Falk B, Garramone S, Shivkumar S. 2004. Diffusion co-

efficient of paracetamol in a chitosan hydrogel. Mater Lett

58:3261–3265.

113. Martin A, Swarbrick J, Cammarata A. 1991. Physical phar-

macy. Mumbai, India: Varghese Publishing House.

114. Gennaro AL. 1990. Remington’s pharmaceutical sciences.

Easton, Pennsylvania: Mack Publishing Company.

115. Costa P, Lobo JMS. 2001. Modeling and comparison of dis-

solution profiles. Eur J Pharm Sci 13:123–133.

116. Mockel JE, Lippold BC. 1993. Zero-order drug release from

hydrocolloid matrices. Pharm Res 10:1066–1070.

117. Ehtezazi T, Washington C, Melia CD. 2000. First order re-

lease rate from porous PLA microspheres with limited exit

holes on the exterior surface. J Control Release 66:27–38.

DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 101, NO. 11, NOVEMBER 2012