Dissolution

113
PREPARED BY : NILESH S.JAWALKAR M.PHARM PHARMACEUTICS

description

DISSOLUTION AND DISSOLUTION TEST APPRATUS PERFECT INFORMATION

Transcript of Dissolution

Page 1: Dissolution

PREPARED BY : NILESH S.JAWALKAR

M.PHARM

PHARMACEUTICS

Page 2: Dissolution

LIST OF CONTENT Introduction Importance and applications Theories of Drug Dissolution Factors affecting Drug Dissolution Various official dissolution test apparatus Dissolution of newer dosage form by

unconventional method and equipment Question bank References

2

Page 3: Dissolution

INTRODUCTION DEFINATION :• Dissolution rate may be defined

as amount of drug substance that goes in the solution per unit time under standard conditions of liquid/solid interface, temperature and solvent composition.

• It can be consider as a specific type of certain hetrogenous reaction in which mass transfer results as a net effect between escape and deposition of solute molecule at a solid surface.

3

Page 4: Dissolution

The processes involved in dissolution of solid dosage forms:

4

Page 5: Dissolution

MECHAMISM OF DISSOLUTION

Initial mechanical lag

Wetting of dosage form

Penetration of dissolution medium

Disintegration Deaggregation Dissolution Occlusion of

some particles

5

Page 6: Dissolution

Intrinsic dissolution rate2 Intrinsic dissolution rate (IDR), which is

the rate of mass transfer per area of dissolving surface and typically has the units of mg cm-2 min-1.

IDR should be independent of boundary layer thickness and volume of solvent. Thus IDR measures the intrinsic properties of the drug only as a function of the dissolution medium, e.g. its pH, ionic strength, counters ions etc.

6

Page 7: Dissolution

Rate of dissolution

Where: m - Amount of dissolved material, kg t - Time, seconds A - Surface area of the interface between the dissolving

substance and the solvent,m2

D - Diffusion coefficient, m2/s d - Thickness of the boundary layer of the solvent at the

surface of the dissolving substance, m Cs - concentration of the substance on the surface,

kg/m3 Cb - concentration of the substance in the bulk of the

solvent, kg/m3

7

Page 8: Dissolution

Importance and Application3

Importance: 1. Results from in-vitro dissolution rate experiment

can be used to explain the observed difference in invivo availability

2. Dissolution testing provides the means to evaluate critical parameters such as adequate bioavailability and provides information necessary to formulator in development of more efficacious and therapeutically optimal dosage forms.

3. Most sensitive and reliable predictors of in-vivo availability.

8

Page 9: Dissolution

4. Dissolution analysis of pharmaceutical dosage forms has emerged as single most important test that will ensure quality of product.

5. It can ensure bioavailability of product between batches that meet dissolution criteria.

6. Ensure batch-to-batch quality equivalence both in-vitro and in-vivo, but also to screen formulations during product development to arrive at optimally effective products.

7. Physicochemical properties of model can be understood needed to mimic in-vivo environment.

9

Page 10: Dissolution

8. Such models can be used to screen potential drug and their associated formulations for dissolution and absorption characteristics.

9. Serve as quality control procedures, once the form of drug and its formulation have been finalized.

10

Page 11: Dissolution

11

Application:2

Page 12: Dissolution

PRODUCT DEVELOPMENT Important tool during development of dosage form. Aids in guiding the selection of prototype

formulations and for determining optimum levels of ingredients to achieve drug release profiles, particularly for extended release formulations.

Also guides in selection of a “market-image” product to be used in pivotal in-vivo bioavailability or bioequivalence studies.

QUALITY ASSURANCE D.T. performed on future production lots and is

used to assess the lot-to-lot performance characteristics of drug product and provide continued assurance of product integrity/similarity.

12

Page 13: Dissolution

PRODUCT STABILITY In-vitro dissolution also used to assess drug

product quality with respect to stability and shelf-life.

As product age, physicochemical changes to the dosage form may alter dissolution characteristics of drug product over time. For some products, polymorph transformations to more stable, and hence less soluble crystalline forms may result in reduced dissolution rates.

13

Page 14: Dissolution

COMPARABILITY ASSESSMENT Also useful for assessing the impact of pre- or

post- approval changes to drug product such as changes to formulation or manufacturing process. Thus, in-vitro comparability assessment is critical to ensure continued performance equivalency and product similarity.

WAIVERS OF IN-VIVO BIOEQUIVALENCE REQUIREMENTS

In-vitro dissolution testing or drug release testing may be used for seeking waiver of required product to conduct in-vivo bioavailability or bioequivalence studies.

14

Page 15: Dissolution

Theories Of Dissolution4

I. Diffusion layer model/Film Theory

II. Danckwert’s model/Penetration or surface renewal Theory

III. Interfacial barrier model/Double barrier or Limited solvation theory

15

Page 16: Dissolution

I. Diffusion layer model/Film Theory It is a simplest model where dissolution of crystal,

immersed in liquid takes place without involving reactive or electrical forces. Consist of two consecutive steps:

1. Solution of the solid to form a thin film or layer at the solid / liquid interface called as stagnant film or diffusion layer which is saturated with the drug this step is usually rapid (instantaneous).

2. Diffusion of the soluble solute from the stagnant layer to the bulk of the solution this step is slower and is therefore the rate determining step in the drugdissolution. The model is depicted in following

fig.16

Page 17: Dissolution

17

Page 18: Dissolution

The rate of dissolution is given by Noyes and Whitney:

Where,dc/dt= dissolution rate of the drugK= dissolution rate constant

Cs= concentration of drug in stagnant layer

Cb= concentration of drug in the bulk of the solution at time t

18

dc dt

k (Cs- Cb)

Page 19: Dissolution

Brunner & Tolloczko incorporated surface area „A‟ in Noyes & Whitney equation.

dc/dt = k1A ( Cs – Cb )

Afterwards Brunner, incorporated Fick‟s law of diffusion & expanded his given eq to include diffusion coefficient „D‟, thickness of stagnant diffusion layer „h‟ & volume of dissolution medium „v‟.

19

Page 20: Dissolution

dc/dt =DAKw/o (Cs- Cb) /Vh

Where, D= diffusion coefficient of drug. A= surface area of dissolving solid. Kw/o= water/oil partition coefficient of drug. V= volume of dissolution medium. h= thickness of stagnant layer. (Cs – Cb )= conc. gradient for diffusion of drug.

20

Modified Noyes & Whitney equation

Page 21: Dissolution

This eq describes a first – order dissolution kinetics. It represents dissolution under non-sink conditions.

If volume is relatively large such that Cs>>>Cb so, dc/dt =AKw/o Cs/Vh Cs & D are constant for each specific chemical

substance so dc/dt =k1 A /Vh

(k1= Kw/o D Cs) V & A are kept constant during dissolution so dc/dt =k

21

Page 22: Dissolution

Sink conditionA Sink conditions describe a dissolution system that is sufficiently dilute so that the dissolution process is not impeded by approach to saturation of the compound of interest.

Sink conditions affect the production of the sample but not the condition of the solution upon sampling.

In vivo condition, there is no conc. build up in the bulk of the solution and hence no retarding effect on the dissolution rate of the drug i.e. Cs>>Cb and sink condition maintain.

22

Page 23: Dissolution

Dissolution rate under sink condition follow zero order dissolution rate.

23

Co

nc

of d

issl

ove

dru

g

Time

First order under non sink condition

Zero order dissolution

Under sink condition

Page 24: Dissolution

For obtaining IVIVC sink condition can be achieved by:

1) Bathing the dissolving solid in fresh solvent from time to time.

2) Increasing the volume of dissolution fluid.3) Removing the dissolved drug by partitioning it

from the aqueous phase of dissolution fluid into the organic phase placed either above or below the dissolution fluid for e.g. hexane or chloroform.

4) Adding a water miscible solvent such as alcohol to the dissolution fluid.

5) By adding selected adsorbents to remove the dissolution drug.

In vitro sink condition is so maintain that Cb always less than 10% of Cs.

24

Page 25: Dissolution

HIXON-CROWELL CUBE ROOT RELATIONSHIP Major assumptions in Noyes-Whitney relationship is that the S.A.

(A) term remains constant throughout dissolution process. This is true for some formulations, such as transdermal patches.

However, size of drug particles from tablets, capsules and suspensions will decrease as drug dissolves.

This decrease in size of particles changes the effective S.A. Thus, Hixon & Crowell modified the eq to represent rate of

appearance of solute by weight in solution by multiplying both sides of volume term.

W01/3– W1/3 = kt W0 = original mass of drug W = mass of drug remaining to dissolve at time t K = dissolution rate constant

25

Page 26: Dissolution

DANCKWERT’S MODEL (PENETRATION OR SURFACE RENEWALTHEORY)

This theory assumes that solid-soln equilibrium is achieved at interface and mass transport is slow step in dissoln process.

The model could be visualized as a very thin film having a conc. Ci which is less than saturation, as it is constantly being exposed to fresh surfaces of liquid having a conc. much less than Ci. Acc. to model, the agitated fluid consist of mass of eddies or packets that are continuously being exposed to new surfaces of solid and then carried back to bulk of liquid.

Diffusion occurs into each of these packets during short time in which the packet is in contact with surface of solid.

Since turbulence actually extends to surface, there is no laminar boundary layer and so no stagnant film exists. Instead, surface continually being replaced with fresh liquid.

26

Page 27: Dissolution

27

Page 28: Dissolution

Interfacial barrier model (double barrier or limited salvation theory) Based on salvation mechanism & solubility rather than diffusion. When considering the dissolution of the crystal will have a different interfacial barrier given by following equation,

G = ki (Cs – Cb) Where G = dissolution per unit area Ki = effective interfacial transport constant

In this theory, the diffusivity D may not be independent of saturation conc. Cs .The interfacial barrier model can be extended to both diffusion layer model and the Dankwert’s model.

28

Page 29: Dissolution

FACTORS AFFECTING DISSOLUTION RATE4

Factors related to Physicochemical Properties of Drug

Factors related to Drug Product Formulation Processing Factor Factors Relating Dissolution Apparatus

Factors Relating Dissolution Test Parameters Miscellaneous factors

29

Page 30: Dissolution

FACTORS RELATED TOPHYSICOCHEMICAL PROPERTIES OF DRUG

1) DRUG SOLUBILITY Solubility of drug plays a prime role in controlling its

dissolution from dosage form. Aqueous solubility of drug is a major factor that determines its dissolution rate. Minimum aqueous solubility of 1% is required to avoid potential solubility limited absorption problems.

Studies of 45 compound of different chemical classes and a wide range of solubility revealed that initial dissolution rate of these substances is directly proportional to their respective solubility.

Ex. Poorly soluble drug :griseofulvin, spironolactone

hydrophilic drug :neomycin30

Page 31: Dissolution

2 ) SALT FORMATION It is one of the common approaches used to increase drug solubility

and dissolution rate. It has always been assumed that sodium salts dissolve faster than their corresponding insoluble acids. Eg.sodium and potassium salts of Peniciilin G, sulfa drugs, phenytoin, barbiturates etc.

While in case of Phenobarbital dissolution of sodium salt was slower than that of weak acid. Same is the case for weak base drug, strong acid salts, such as hydrochlorides and sulphates of weak bases such as epinephrine, tetracycline are commonly used due to high solubility. However, free bases of chlortetracycline, methacycline were more soluble than corresponding hydrochloride salt at gastric pH values, due to common ion suppression.

31

Page 32: Dissolution

3) PARTICLE SIZE There is a direct relationship between surface area of drug and its

dissolution rate. Since, surface area increases with decrease in particle size, higher dissolution rates may be achieved through reduction of particle size.

Micronization of sparingly soluble drug to reduce particle size is by no means a guarantee of better dissolution and bioavailability.

Micronization of hydrophobic powders can lead to aggregation and floatation. when powder is dispersed into dissolution medium. So, mere increase in S.A. of drug does not always guarantee an equivalent increase in dissolution rate. Rather, it is increase in the “effective” S.A., or area exposed to dissolution medium and not the absolute S.A. that is directly proportional to dissolution rate.

Hydrophobic drugs like phenacetin, aspirin shows decrease in dissoln. rate as they tend to adsorb air at the surface and inhibit their wettability. Problem eliminated by evacuating surface from adsorbed air or by use of surfactants. So these drugs in-vivo exhibit excellent wetting due to presence of natural surfactants such as bile salts

Eg. therapeutic conc. of griseofulvin was reduced to half by micronization

32

Page 33: Dissolution

4) SOLID STATE CHARACTERISTICS Solid phase characteristics of drug, such as amorphicity,

crystallinity, state of hydration and polymorphic structures have significant influence on dissolution rate.

Anhydrous forms dissolve faster than hydrated form because they are thermodynamically more active than hydrates. Eg. Ampicillin anhydrate faster dissolution rate than trihydrate.

Amorphous forms of drug tend to dissolve faster than crystalline materials. E.g.Novobiocin suspension, Griseofulvin.

Where in the dissolution rate of amorphous erythromycin estolate is markedly lower than the crystalline form of erythromycin estolate.

Metastable(high activation energy)polymorphic form have better dissolution than stable form

33

Page 34: Dissolution

5) Co precipitation &/or Complexation

Co precipitation as well as complexation are use for enhancing the dissolution rate of drug due to,

Formation energetic amorphous drug phase or Drug being molecularly dispersed orFormation of co accervates

e.g.1) Hydroflumethiazide – PVP co precipitate has four times more solubility than crystalline drug.

2) Dissolution rate of sulfathiazole could be significantly increased by co precipitating the drug with povidone

34

Page 35: Dissolution

Factors related to Drug Product Formulation

1)DILUENTS Studies of starch on dissolution rate of salicylic acid

tablet by dry double compression process shows three times increase in dissolution rate when the starch content increase from the 5 – 20 %.

Here starch particles form a layer on the outer surface of hydrophobic drug particles resulting in imparting hydrophilic character to granules & thus increase in effective surface area & rate of dissolution

35

Page 36: Dissolution

Time in min.

10 20 30 40 50

100

80

60

40

20Am

t of

dis

solv

ed m

g

10% starch

5% starch

The dissolution rate is not only affected by nature of the diluent but also affected by excipient dilution (drug/excipient ratio).E.g. in quinazoline comp. dissolution rate increases as theexcipient /drug ratio increases from 3:1 to 7:1 to 11:1.

36

Page 37: Dissolution

2)DISINTEGRANTS Disintegrating agent added before & after the granulation affects the

dissolution rate. Studies of various disintegrating agents on Phenobarbital tablet

showed that when copagel (low viscosity grade of Na CMC) added before granulation decreased dissolution rate but if added after did not had any effect on dissolution rate.

Microcrystalline cellulose is a very good disintegrating agent but at high compression force, it may retard drug dissolution.

Starch is not only an excellent diluent but also superior disintegrant due to its hydrophilicity and swelling property.

3)BINDERS AND GRANULATING AGENTS The hydrophilic binder increase dissolution rate of poorly wettable

drug. Large amt. of binder increase hardness & decrease disintegration

/dissolution rate of tablet. Non aqueous binders such as ethyl cellulose also retard the drug

dissolution.37

Page 38: Dissolution

Phenobarbital tablet granulated with gelatin solution provide a faster dissolution rate in human gastric juice than those prepared using Na –carboxymethyl cellulose or polyethylene glycol 6000 as binder.

In Phenobarbital tablet, faster dissolution rate was observed with 10% gelatinwhereas decrease in dissolution rate with 20% gelatin. This was due to higherconcentration which formed a thick film around tablet.

Water soluble granulating agent Plasdone gives faster dissolution rate comparedto gelatin.

38

Page 39: Dissolution

4) Lubricants

Lubricants are hydrophobic in nature (several metallic stearate & waxes) which inhibit wettability, penetration of water into tablet so decrease in disintegration and dissolution.

The use of soluble lubricants like SLS and Carbowaxes which promote drug dissolution.

39

Page 40: Dissolution

5)SURFACTANTS They enhance the dissolution rate of poorly soluble drug. This is due to

lowering of interfacial tension, increasing effective surface area, which in turn results in faster dissolution rate.

E.g. Non-ionic surfactant Polysorbate 80 increase dissolution rate of

phenacetin granules.6)WATER-SOLUBLE DYES Dissolution rate of single crystal of sulphathiazole was found to decrease

significantly in presence of FD&C Blue No.1. The inhibiting effect was related to preferential adsorption of dye

molecules on primary dissolution sources of crystal surfaces. They inhibit the micellar solubilization effect of bile salts on drug.

Riboflavin tablet decrease when used FD & C Red no.3 dye in film coat

7)Effect of coating component on tablet dissolution

Coating ingredient especially shellac & CAP etc. Also have significant effect on the dissolution rate of coated tablet. Tablets with MC coating were found to exhibit lower dissolution profiles than those coated with HPMC at 37ºC.

40

Page 41: Dissolution

PROCESSING FACTORS

1) METHOD OF GRANULATION

Granulation process in general enhances dissolution rate of poorly soluble drug.

Wet granulation is traditionally considered superior. But exception is the dissolution profile of sodium salicylate tablets prepared by both wet granulation and direct compression where the dissolution was found more complete and rapid in latter case.

A newer technology called as APOC “Agglomerative Phase of Comminution” was found to produce mechanically stronger tablets with higher dissolution rates than those made by wet granulation. A possible mechanism is increased internal surface area of granules produced by APOC method.

41

Page 42: Dissolution

2)COMPRESSION FORCE The compression process influence density, porosity, hardness,

disintegration time & dissolution of tablet.

42

1. tighter bonding

2 . higher compression force cause deformation crushing or fracture of drug particle or convert a spherical granules into disc Shaped particle

3.& 4. both condition

Page 43: Dissolution

3) DRUG EXCIPIENT INTERACTION These interactions occur during any unit operation such as mixing,

milling ,blending, drying, and/or granulating result change in dissolution.

The dissolution of prednisolone found to depend on the length of mixing time with Mg-stearate

Similar as increase in mixing time of formulation containing 97 to 99% microcrystalline cellulose or another slightly swelling disintegrant result in enhance dissolution rate.

Polysorbate-80 used as excipient in capsules causes formation of formaldehyde by autoxidation which causes film formation by denaturing the inner surface of capsule. This causes decrease in dissoln rate of capsules.

4) STORAGE CONDITIONS Dissolution rate of hydrochlorothiazide tablets granulated with

acacia exhibited decrease in dissolution rate during 1 yr of aging at R.T

For tablets granulated with PVP there was no change at elevated temperature but slight decrease at R.T.

Tablets with starch gave no change in dissoln. rate either at R.T. or at elevated temperature.

43

Page 44: Dissolution

FACTORS RELATING DISSOLUTION APPARATUS1

1) AGITATION Relationship between intensity of agitation and rate of

dissolution varies considerably acc. to type of agitation used, the degree of laminar and turbulent flow in system, the shape and design of stirrer and physicochemical properties of solid.

Speed of agitation generates a flow that continuously changes the liq/solid interface between solvent and drug. In order to prevent turbulence and sustain a reproducible laminar flow, which is essential for obtaining reliable results, agitation should be maintained at a relatively low rate.

Thus, in general relatively low agitation should be applied.

I. BASKET METHOD- 100 rpm II. PADDLE METHOD- 50-75 rpm

44

Page 45: Dissolution

2) STIRRING ELEMENT ALIGNMENT

The USP / NF XV states that the axis of the stirring element must not deviate more than 0.2 mm from the axis of the dissolution vessel which defines centering of stirring shaft to within ±2 mm.

Studies indicant that significant increase in dissolution rate up to 13% occurs if shaft is offset 2-6 mm from the center axis of the flask.

Tilt in excess of 1.5 0 may increase dissolution rate from 2 to 25%.

3) SAMPLING PROBE POSITION & FILTER Sampling probe can affect the hydrodynamic of the system & so that

change in dissolution rate. For position of sampling, USP states that sample should be removed

at approximately half the distance from the basket or paddle to the dissolution medium and not closer than 1 cm to the side of the flask.

Filter material must be saturated with the drug by repeated passage to avoid losses that might go undetected during the test sampling.

Accumulation of the particulate matter on the surface may cause significant error in the dissolution testing.

45

Page 46: Dissolution

FACTORS RELATING DISSOLUTION TEST PARAMETERS

1)TEMPERATURE Drug solubility is temperature dependent, therefore careful

temperature control during dissolution process is extremely important.

Generally, a temp of 37º ± 0.5 is maintained during dissolution of oral dosage forms and suppositories. However, for topical preparations temp as low as 30º and 25º have been used

2) DISSOLUTION MEDIUM Effect of dissolution air on dissolution medium Altering PH Dissolved air tends to release slowly in form of tiny air bubble that

circulate randomly and affect hydrodynamic flow pattern Specific gravity decrease thus floating of powder thus wetting and

penetration problem. Dissolution media composition & PH Addition of Na – sulfate decrease the dissolution rate. Addition of urea increase dissolution rate. 46

Page 47: Dissolution

Volume of dissolution medium and sink conditions  Volume generally 500, 900 or 1,000 ml. Simulated gastric fluid(SGF) - pH 1.2. Simulated intestinal fluid (SIF)- pH 6.8 (not exceed pH 8.0). The need for enzymes should be evaluated case-by-case like….

(Pepsin with SGF and pancreatin with SIF If drug is poorly soluble, a relatively large amount of fluid should be

used if complete dissolution is to be expected. In order to minimize the effect of conc. gradient and maintain sink

conditions, the conc. of drug should not exceed 10-15% of its max. Solubility in dissoln. medium selected. For most of the drugs about 1 L is more than sufficient to maintain sink conditions.

However, some insoluble drug present a problem as to handling of huge volume of dissoln. medium that would be required to maintain the sink conditions. For these, different approaches have been tried like…. 

continous flow method where fresh solvent is pumped continuously into dissoln flask at a fixed flow rate while maintaining a constant volume.

Use of non-ionic surfactant in conc. above CMC. Use of alcoholic solution (10-30%).

47

Page 48: Dissolution

Miscellaneous factor

1. Absorption :

Absorbent increase the dissolution rate under condition of a decrease concentration gradient applying Nerst–Brunner film theroy

2. Humidity : Moisture has been shown to influence

the dissolution of many drug from solid dosage form.

48

Page 49: Dissolution

Dissolution Test Apparatus1

I.P. I.P. USPUSP B.P.B.P. E.P. E.P.

Type 1Type 1Paddle Paddle

apparatusapparatus Basket Basket apparatusapparatus

Basket Basket apparatusapparatus

Paddle Paddle apparatusapparatus

Type 2Type 2Basket Basket apparatusapparatus

Paddle Paddle apparatusapparatus

Paddle Paddle apparatusapparatus

Basket Basket apparatusapparatus

Type 3Type 3Reciprocating Reciprocating cylindercylinder

Flow through cell Flow through cell apparatusapparatus

Flow through Flow through cell apparatuscell apparatus

Type 4Type 4Flow through Flow through cell apparatuscell apparatus

Type 5Type 5Paddle over Paddle over diskdisk

Type 6Type 6 cylindercylinder

Type 7Type 7Reciprocating Reciprocating holderholder 49

Page 50: Dissolution

Solid dosage form (tablet & capsule)I.P. & E.P.

Apparatus I – paddle apparatus Apparatus II – basket apparatusB.P. & U.S.P. Apparatus I – basket apparatus Apparatus II – paddle apparatusB.P. & E.P. Apparatus III – flow through cell apparatusConditions ( for all)

Temp. - 37±0.50C PH - ±0.05 unit in specified monograph Capacity – 1000 ml Distance between inside bottom of vessel and paddle/basket is

maintained at 25±2 mm. For enteric coated dosage form it is first dissolved in 0.1 N HCl &

then in buffer of pH 6.8 to measure drug release. (Limit – NMT 10% of drug should dissolve in the acid after 2hr.and about 75% of it should dissolve in the buffer after 45 min.

50

Page 51: Dissolution

USP APP. USP APP. DESCRIPTOINDESCRIPTOIN ROT. ROT. SPEEDSPEED

DOSAGE DOSAGE FORMFORM

Type 1Type 1 Basket apparatusBasket apparatus 50-120rpm50-120rpm IR, DR, ERIR, DR, ER

Type 2Type 2 Paddle apparatusPaddle apparatus 25-50rpm25-50rpm IR, DR, ERIR, DR, ER

Type 3Type 3 Reciprocating Reciprocating cylindercylinder

6-35rpm6-35rpm IR,ERIR,ER

Type 4Type 4 Flow through cell Flow through cell apparatusapparatus

N/AN/A ER , poorly ER , poorly soluble APIsoluble API

Type 5Type 5 Paddle over diskPaddle over disk 25-50rpm25-50rpm TRANSDERMALTRANSDERMAL

Type 6Type 6 cylindercylinder N/AN/A TRANSDERMALTRANSDERMAL

Type Type Reciprocating Reciprocating holderholder

30rpm30rpm ERER

51

Page 52: Dissolution

APPARATUS 1- BASKET APPARATUS

Dosage form contained within basket

Dissolution should occur within Basket

Useful for : Tablets Capsules –Beads –Floaters

pH change by media exchange

52

Page 53: Dissolution

• Drug product – Solids (mostly floating) • Monodisperse (tablets) • Polydisperse (encapsulated beads)• Agitation – Rotating stirrer – Usual speed: 50 to 100 rpm• Disadvantage– Formulation may clog to 40 mesh screen

53

Page 54: Dissolution

USP Apparatus 2 – Paddle Dosage form should

remain at the bottom centre of the vessel

Sinkers used for floaters

Useful for : – Tablets – Capsules pH change by media

addition

54

Page 55: Dissolution

• Drug product – Solids (mostly non floating) • Monodisperse (tablets) • Polydisperse (encapsulated beads)

• Agitation – Rotating stirrer – Usual speed: 25 to 75 rpm

Standard volume: 900/1000 ml Advantages:1. Easy to use and robust2. Ph change possible 3. Can be easily adapted to apparatus 5

Disadvantages – Floating dosage forms require sinker – Positioning of tablet 55

Page 56: Dissolution

56

Page 57: Dissolution

Limitations of USP Apparatus 1and 2:1. USP2 (and USP1) Apparatus has plenty of

HYDRODYNAMICS.

2. Complicated 3-dimensional flow generated by the paddle.

3. Significant impact of convective transport –Conditions used (50 – 100 rpm) highly exaggerates flow in the GI.

4. Use of solvents and surfactants non-native to GI.

57

Page 58: Dissolution

Apparatus III – Reciprocating cylinder

The assembly consists of a 1)set of cylindrical, 2)flat-bottomed glass vessels;

a 3)set of glass reciprocating cylinders;

4)stainless steel fittings (type 316 or equivalent) and screens that are made of suitable nonsorbing and nonreactive material(polypropelene) and that are designed to fit the tops and bottoms of the reciprocating cylinders; and a motor and drive assembly to reciprocate the cylinders vertically inside the vessels 58

Page 59: Dissolution

The vessels are partially immersed in a suitable water bath of any convenient size that permits holding the temperature at 37 ± 0.5 during the test.

The dosage unit is placed in reciprocating cylinder & the cylinder is allowed to move in upward and downward direction constantly. Release of drug into solvent within the cylinder measured.

Useful for: Tablets, Beads, controlled release formulations

Standard volume: 200-250 ml/station

Advantages: 1) Easy to change the pH-profiles 2) Hydrodynamics can be directly influenced by varying

the dip rate.

Disadvantages: 1) small volume (max. 250 ml) 2) Little experience 3) Limited data

59

Page 60: Dissolution

Apparatus 3 – Reciprocating cylinder

60

Page 61: Dissolution

USP Apparatus 4 - Flow Through Cell

61

Page 62: Dissolution

The assembly consists of a reservoir and a pump for the Dissolution Medium; a flow-through cell; a water bath that maintains the Dissolution Medium at 37 ± 0.5

The pump forces the Dissolution Medium upwards through the flow-through cell.

Assemble the filter head, and fix the parts together by means of a suitable clamping device.

Introduce by the pump the Dissolution Medium warmed to 37 ± 0.5 through the bottom of the cell to obtain the flow rate specified in the individual monograph.

Collect the elute by fractions at each of the times stated. Perform the analysis as directed in the individual

monograph

62

Page 63: Dissolution

Useful for: Low solubility drugs, Micro particulates, Implants, Suppositories, Controlledrelease formulations

Variations: (A) Open system & (B) Closed system Advantages: 1. Easy to change media pH2. PH-profile possible 3. Sink conditions Disadvantages: 1. Deaeration necessary 2. High volumes of media 3. Labor intensive

63

Tablets 12 mm Tablets 22,6 mm Powders / Granules Implants Suppositories / Soft gelatine capsules

Page 64: Dissolution

Apparatus 4 – Flow-Through Cell

64

Page 65: Dissolution

USP Apparatus 5 - Paddle Over Disk

65

Page 66: Dissolution

Use the paddle and vessel assembly from Apparatus 2 with the addition of a stainless steel disk assembly designed for holding the transdermal system at the bottom of the vessel.

The disk assembly holds the system flat and is positioned such that the release surface is parallel with the bottom of the paddle blade

The vessel may be covered during the test to minimize evaporation. Useful for: Transdermal patches

Standard volume: 900 ml Disadvantages: Disk assembly restricts the patch size. Borosilicate Glass 17 mesh is standard (others available) Accommodates patches of up to 90mm 66

Page 67: Dissolution

USP Apparatus 6 - Cylinder

67

Page 68: Dissolution

• Use the vessel assembly from Apparatus 1 except to replace the basket and shaft with a stainless steel cylinder stirring element

• The temperature is maintained at 32°C ± 0.5°C

• The dosage unit is placed on the cylinder with release side out

The dosage unit is placed on the cylinder at the beginning of each test, to the exterior of the cylinder such that the long axis of the system fits around the circumference of the cylinder & removes trapped air bubbles.

Place the cylinder in the apparatus, and immediately rotate at the rate specified in the individual monograph.

68

Page 69: Dissolution

USP Apparatus 7 – Reciprocating Holder

69

Page 70: Dissolution

• The assembly consists of a set of volumetrically calibrated solution containers made of glas or other suitable inert material, a motor and drive assembly to reciprocate the system vertically

• The temperature is maintained at 32°C ± 0.5°C • The dosage unit is placed on the cylinder with release side out The

solution containers are partially immersed in a suitable water bath of any convenient size that permits maintaining the temperature, inside the containers at 32 ± 0.5 For Coated tablet drug delivery system attach each system to be tested to a suitable Sample holder

70

Page 71: Dissolution

•For Transdermal drug delivery system attach the system to a suitable sized sample holder with a suitable O-ring such that the back of the system is adjacent to and centered on the bottom of the disk-shaped sample holder or centered around the circumference of the cylindrical-shaped sample holder. Trim the excess substrate with a sharp blade.

71

Page 72: Dissolution
Page 73: Dissolution

Novel Dissolution test Apparatus for buccal and sublingual tablets. 13

Buccal and sublingual dissolution differs from g.i.dissolution in following ways…

smaller volume ( of saliva) short residence time ( in mouth) solids transfer composition of fluid ( saliva composition) Incomplete dissolution

So, our dissolution apparatus must provide above conditions for performing dissolution test of buccal and sublingual tablets.

73

Page 74: Dissolution

MODEL:-IIt provides all the characteristics described above.It is given by HUGHES

6ml/min

74

Page 75: Dissolution

This novel system comprises a single stirred continuous flow-through cell that includes …

a dip tube

a central shaft with propeller &

a filter along with one inlet for saliva & one outlet for sample.

A pump is available which pushes the fluid at the rate 6 ml/min. and this will give a residence time in the cell of approximately 100 secs for 63% of the dosage form & gives complete removal in about 8 mins.

75

Page 76: Dissolution

Composition of saliva:-

Composition of stimulated salivaComposition of stimulated saliva

KHKH22POPO44 12mM12mM

NaClNaCl 4040mMmM

CaClCaCl22 1.51.5mMmM

NaOHNaOH To pH 6.2 To pH 6.2

76

Page 77: Dissolution

COMPOUNDCOMPOUND mMmM

CaClCaCl22. 2H. 2H22OO 0.20.2

MgClMgCl22 . 6H . 6H22OO 0.0610.061

NaCl NaCl 1.0171.017

KK22COCO33 . 5H . 5H22OO 0.6030.603

NaNa22HPOHPO44 .7H .7H22OO 0.200.20

NaNa22HPOHPO44.H.H22OO 0.2730.273

Submaxillary fluidSubmaxillary fluid 1.01.0

α amylaseamylase 2.02.0

Composition of sublingual saliva

77

Page 78: Dissolution

Advantage It is a rapid, taking only about 20 minutes per test &

repeatable. This method could be used as a QC test to ensure

dosage uniformity. This method is particularly suited for evaluating taste

masking. Application This dissolution apparatus is used for -Claritin -Reditabs -Zydis system

78

Page 79: Dissolution

MODEL:-II International Journal of Pharmaceutics ,October 2006

The device introduced by them is based on the circulation of pre-warmed dissolution medium through a cell.

Buccal tablet was attached on chicken pouches.

They stated “ the results obtained by this using this apparatus for the release of drug from bio adhesive tablets concurred with the predicted patterns.”

79

Page 80: Dissolution

Method for dissolution testingMethod for dissolution testing

In vitro target site – animal buccal tissues or buccal cell culturesIn vitro target site – animal buccal tissues or buccal cell cultures

Animal sacrificed just before in vitro testing

Buccal mucosa removed surgically

cold Krebs buffer Isolated buccal mucosa stored in ice

Mounted between side by side diffusion cells

80

Page 81: Dissolution

81

Novel Dissolution test Apparatus for Floating tablets8

Ideal qualities for dissolution apparatus for Floating tablets.

I. Dosage form should not stick on the agitating device. Therefore, under driven arrangement is more suitable.II. The test must try to mimic the gastric juice release

rate (2-4ml/min).iii) The sample collection must be easy.iv) The volume of cell having dosage form in it must have

nearly same volume as compared to in-vivo gastric volume.

(70 ml).

Page 82: Dissolution

MODIFIED ROSSET –RICE TEST

82

Page 83: Dissolution

Better in-vivo-in-vitro correlation shown by this method.

It mimics three points. i) gastric volume ii) gastric acid secretion iii) gastric emptying. which USP-II apparatus fails to

mimic. &Here, tablet does not stick to agitating device

because it is under-driven.

83

Page 84: Dissolution

Dissolution study of Lipid –filled soft

gelatin capsules. 15

84

(International Journal of Pharmaceutics ,October 2006.)

Page 85: Dissolution

MECHANISM It is one type of flow through cell.

Lipid content due to its lower density rises up in the cell after rupturing of the capsule.

When lipid phase reaches the triangular area top of the left side cell, it stays there.

thus ,dissolution medium continuously extracts the drug from the lipid layer as it flows through the cell.

The dissolved drug can now be determined using a fractional collector and be analyzed in the medium.

85

Page 86: Dissolution

Pillay & Fassihi model for Lipid –filled soft gelatin capsules.

86

I = organic phase, i.e., 100 ml II = aqueous phase III = ring/mesh assembly

IV = position of capsule

Page 87: Dissolution

Dissolution study of chewing gum as a dosage form. 11

87

Page 88: Dissolution

European Pharmacopoeia published a monograph describing a suitable apparatus for studying the in vitro release of drug substances from chewing gums.

A study was carried out to explore differences in the release of nicotine from the directly compressible gum base compared with a conventional nicotine gum using the European Pharmacopoeia chewing apparatus described in the European Pharmacopoeia.

88

Page 89: Dissolution

The gums were placed in the chewing chamber with 40 ml of artificial saliva.

The temperature of the chewing chamber :37±1°C chew rate : 60 chews/minute unspecified buffer (with a pH close to 6) : 20 ml

The machine was run without chewing gum for the first two minutes and then the buffer removed to ensure that

i) any residues from the extensive washing and cleaning procedure were removed &

ii) to allow equilibration of chew rate and temperature.

89

Page 90: Dissolution

Artificial Saliva Formulation Components:-

90

Page 91: Dissolution

Dissolution study of the bio-degradable microspheres.

91

Page 92: Dissolution

Mini Paddle Apparatus- for dissolution study of Immediate-Release Dosage

Forms10

92

Page 93: Dissolution

The mini paddle is based on the USP paddle setup but scaled down exactly 1/3 with respect to the dimensions.

250 ml volume used in the mini paddle apparatus.

A stirring rate of 100 rpm in the mini paddle apparatus appears to be the most favorable.

93

Page 94: Dissolution

Mini paddle apparatus might be a useful tool in characterizing drug release profiles under “standard test conditions.”

Due to the possibility of using smaller sample sizes and smaller volumes of media ,

it offers various advantages in terms of substance, analytical, and material cost savings.

The mini paddle set-up is also a promising alternative in the case of highly potent drugs.

The mini paddle should preferably be used for… powders, multiparticulate dosage forms, small tablets or capsules

(i.e., where the paddle apparatus would be the usual method of choice)

94

Page 95: Dissolution

TDDS Dissolution Test12

Apparatus used: Franz diffusion apparatus Paddle over disk Cylinder method Flow through diffusion cell

95

Page 96: Dissolution

Animal skin – hairless mouse , guinea pig , rabbitAnimal skin – hairless mouse , guinea pig , rabbit But no animal skin mimic human skin so human skin is preferred & But no animal skin mimic human skin so human skin is preferred & use is based on availability. use is based on availability. Temp 32c +/- 1c pH 5-6 Stirring rate – 100 Temp 32c +/- 1c pH 5-6 Stirring rate – 100 rpm.rpm. Enzymes for oxidatn, reductn, hydrolys,conjgtnEnzymes for oxidatn, reductn, hydrolys,conjgtnSkin lipidsSkin lipidsMicrobial flora ( difficult to reproduceMicrobial flora ( difficult to reproduce 96

Page 97: Dissolution

Semisolid dissolution test12

Apparatus used USP paddle over disk Franz diffusion apparatus Flow through apparatus Unconventional apparatus a) designed by chouhan12

Commonly used membranes Polysulphone (Tuffryn, 0.45 μm size): Most suitable syn.

membrane for ointments. Cellulosic acetate plus. Nylon Teflon and polycorbonate 97

Page 98: Dissolution

A = Constant temp. water bathB = 250 ml beakerC = Teflon diskD = Layer of ointmentE = sinkF = Magnetic stirring barG = Motor

98

Page 99: Dissolution

99

Page 100: Dissolution

Parentral Depot dissolution test14

IN VITRO DISSOLUTION TESTING FOR DEPOTS

DEPOT

Dissolution

DRUG IN SOLUTION

Partition

DRUG IN TISSUE FLUID

Factors need to be controlled.

1) nature & site of absorption of drug

2) physiological pH of tissue fluid

100

Page 101: Dissolution

DIAGRAM OF ROTATING DIALYSIS CELLDIAGRAM OF ROTATING DIALYSIS CELL

IT USED FOR PARENTERAL DEPOTSDialysis membrane provides well defined surface area.Dissolution medium pH = pH of site of absorption.E.g NAPROXEN – IN COCONUT OILVOLUME – 1000ML , 37C+/_ 0.5CStirring rate 50rpmDissolution media pH 3 ± 0.01 (0.05 M phosphate buffer) pH 5 ± 0.01 (0.05 M Acetate buffer) pH 7 ± 0.01 (0.05 M phosphate buffer) 101

Page 102: Dissolution

Dissolution Test of Suspension15 APPARATUS

Rotating Paddle MEDIA

Aqueous medium

Rotationg speed 50rpm

Temperature 37oc +/- 5

102

Page 103: Dissolution

Method parameters such as sample introduction and agitation rate should be established on the basis of the viscosity and composition of the suspension matrix. .

For low-viscosity suspensions, an accurate dose can be delivered to the bottom of the dissolution vessel using a volumetric pipette.

A slow agitation rate of 25 rpm is generally recommended for less viscous sus-pensions.

For high-viscosity samples, the dose may need to be determined by weight with a quantitative sample transfer to the dissolution vessel to ensure ac-curacy of the sample size introduced.

High-viscosity suspensions may also require a faster agitation rate such as 50 or 75 rpm to prevent sample mounding at the bottom of the vessel. 103

Page 104: Dissolution

Dissolution Test of Inhaler16 A USP Apparatus 2,

Hanson SR8-Plus dissolution test station was employed to conduct the dissolution study.

A schematic diagram of modifications to the dissolution apparatus is shown in Figure 1. The two main components of the dissolution setup include

1) dissolution test station (FigureA)

2) a newly designed membrane holder (Figure B). 104

Page 105: Dissolution

The membrane holder assembly was customized, it consists of

NGI dissolution cup (a),

removable impaction inset (b),

a securing ring (c), two sealing o-rings, a PC membrane to

function as a highly porous diffusional powder retaining layer.

105

Page 106: Dissolution

To select suitable particle size cutoff ranges for the subsequent dissolution study, aerodynamic particle separation was achieved using the NGI. Either the Ventolin HFA device or the Pulmicort Flexhaler device was actuated five times to obtain a quantifiable amount of drug.

The NGI was operated at a flow rate of 30 L/min for the Ventolin HFA and 60 L/min for the Pulmicort Flexhaler.

For the dissolution studies, the dissolution cup assembled with the impaction insert was placed in the NGI, as shown in Figure 2. Following actuation, the impaction insert was removed from the NGI dissolution cup for the dissolution test

106

Page 107: Dissolution

Dissolution Media Simulated lung fluid (SLF), 0.2 M phosphate buffer(pH 7.4), phosphate-buffered saline (PBS), modified PBS(mPBS) containing

dipalmitoylphosphatidylcholine(DPPC), and PBS containing polysorbate 80 (tPBS) were used in the dissolution studies.

107

Page 108: Dissolution

108

Page 109: Dissolution

Dissolution Apparatus

The membrane holder was placed at the bottom of each vessel, release-surface side up, with the distance between the bottom edge of the paddle and the surface of the membrane holder maintained at 20 ± 2 mm( Figure 2)

The distance between the paddle and the surface of the membrane holder can be adjusted within a range that allows the paddle to effectively remove released drug from the exposed membrane surface and provides continuous circulation to the media in the vessel.

109

Page 110: Dissolution

MECHANISMThe mechanism of this dissolution method can be explained by a dissolution–diffusion-controlled drug release from the membrane holder. During the dissolution process, the dispersed drug within the membrane holder undergoes dissolution as dissolution medium migrates through the pores on the membrane surface, and the dissolved drug then releases out to the reservoir by diffusion

DIFFUSION MEMBRANE A PC membrane was selected as the diffusion barrier .

The PC membrane surface constitutes a perfect sink for the released drug when used in this manner.

A PC membrane do not swell, do not create air bubbles, have a well-defined uniform pore size (17, 18), and consist of homogeneous 0.05-μm non-tortuous cylindrical pores on the surface that allow free diffusion of dissolved drug and dissolution medium

110

Page 111: Dissolution

List Of Reference

111

1. Pharmaceutical dissolution testing by Umesh V. Banakar.2. Remington 20th edition, 654.3. Dissolution, Bioavailability & Bioequivalence by M. Abdou.4. Biopharmaceutics & Pharmacokinetics by D.M.Bramankar, 20-29.5. Generic Drug Product Development, vol-143 by Shargel.6. Current concepts in pharmaceutical sciences & biopharmaceutics by James Swarbrick.7. Advanced Drug Delivery Reviews,(46), 75-87, 2001.8.A Novel Multi compartment Dissolution Apparatus for Evaluation of Floating Dosage Form Containing Poorly Soluble Weakly Basic Drug Dr. Rajesh K. Parikh1,2,Dhaivat C. Parikh1,Renish R. Delvadia1, and Sanjay M. Patel Dissolution Technologies ,FEBRUARY 2006.

9.A More Relevant Dissolution Method for Evaluation of Floating Drug Delivery System Mukesh C.Gohel, Pavak R.Mehta, Rikita K. Dave2 and Nehal H. Bariya. Dissolution Technologies | NOVEMBER 2004.

Page 112: Dissolution

10.The Mini Paddle Apparatus–a Useful Tool in the Early Developmental Stage? Experiences with Immediate-Release Dosage Forms. Dissolution Technologies | NOVEMBER 2006.

11.in Vitro Release of Nicotine From Chewing Gum Formulations. Dissolution Technologies | MAY 2004.

12.Current perspectives in dissolution testing of unconventional and novel dosage forms semisolid Shirzad Azarmi, Wilson Roac, Raimar L¨obenberg , International Journal of Pharmaceutics ,October 2006

13.A new method of characterising buccal dissolution of drugs. Dr.L.Hughes.Rohm & Haas research laboratories.feb.2004.

14.FIP/AAPS Guidelines for Dissolution/In Vitro Release Testing of Novel/Special Dosage Forms . Dissolution Technologies | FEBRUARY 2005

15. 14.FIP/AAPS Guidelines for Dissolution/In Vitro Release Testing of Novel/Special Dosage Forms (suspension) march 2010

16.Invitro dissolution testing for inhaler ,dissolution technology ,november 2010

112

Page 113: Dissolution

113