SlideShare a Scribd company logo
1 of 131
DRUG DISSOLUTION
Definition- 
• Dissolution is a process in which a solid 
substance solubilizes in a given solvent i.e. 
mass transfer from the solid surface to the liquid 
phase. 
• Rate of dissolution is the amount of drug 
substance that goes in solution per unit time 
under standardized conditions of liquid/solid 
interface, temperature and solvent composition. 
2
DDrruugg DDiissssoolluuttiioonn PPrroocceessss
• Initial mechanical 
lag 
• Wetting of dosage 
form 
• Penetration of 
dissolution 
medium 
• Disintegration 
• Deaggregation 
• Dissolution 
• Occlusion of some 
particles 
4
 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. 
5
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 - DDiiffffuussiioonn ccooeeffffiicciieenntt, 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 
6
 Application:2 
7
Theories of Drug Dissolution 
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.
I. Diffusion layer model/Film Theory :- 
• It involves two steps :- 
a. Solution of the solid to form stagnant film or 
diffusive layer which is saturated with the drug 
b. Diffusion of the soluble solute from the stagnant 
layer to the bulk of the solution; this is r.d.s in 
drug dissolution.
• The rate of dissolution is given by Noyes and 
Whitney: 
= k (Cs- Cb) dc 
dt 
Where, 
dc/dt= dissolution rate of the drug 
K= dissolution rate constant 
Cs= concentration of drug in stagnant layer 
Cb= concentration of drug in the bulk of the 
solution at time t
Modified Noyes-Whitney’s Equation - 
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.
Sink condition 
A 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. 
13
• Dissolution rate under sink condition follow zero 
order dissolution rate. 
Conc of disslove drug 
Time 
Zero order dissolution 
Under sink condition 
First order under non 
sink condition 
14
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. 
15
 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 16
• This is first order dissolution rate process, for 
which the driving force is concentration gradient. 
• This is true for in-vitro dissolution which is 
characterized by non-sink conditions. 
• The in-vivo dissolution is rapid as sink conditions 
are maintained by absorption of drug in systemic 
circulation i.e. Cb=0 and rate of dissolution is 
maximum.
• Under sink conditions, if the volume and surface 
area of the solid are kept constant, then 
dC 
dt 
= K 
• This represents that the dissolution rate is 
constant under sink conditions and follows zero 
order kinetics.
II. Danckwert’s model/Penetration or 
surface renewal Theory :- 
• Dankwert takes into account the eddies or 
packets that are present in the agitated fluid 
which reach the solid-liquid interface, absorb 
the solute by diffusion and carry it into the bulk 
of solution. 
• These packets get continuously replaced by 
new ones and expose to new solid surface 
each time, thus the theory is called as surface 
renewal theory.
• The Danckwert’s model is expressed by 
equation 
dC 
dt = dm 
V = A (Cs-Cb). γ D 
dt 
Where, 
m = mass of solid dissolved 
Gamma (γ) = rate of surface renewal
III. Interfacial barrier model/Double barrier or 
Limited solvation theory :- 
• The concept of this theory is explained by 
following equation- 
G = Ki (Cs - Cb) 
Where, 
G = dissolution rate per unit area, 
Ki = effective interfacial transport constant.
3) Interfacial layer model 
S 
Film boundary 
Bulk solution 
Cs 
C 
Stagnant layer 
In this model it is assumed that the reaction at solid surface is not 
instantaneous i.e. the reaction at solid surface and its diffusion across the 
interface is slower than diffusion across liquid film. 
therefore the rate of solubility of solid in liquid film becomes the rate 
limiting than the diffusion of dissolved molecules 
equation : dm/dt = Ki (Cs – C ) K = effective interfacial transport rate 
constant
Biopharmaceutical Classification System 
High Solubility 
(Dose Vol. NMT 
250 mL) 
Low Solubility 
(Dose Vol. >250 
mL) 
High Permeability 
(Fract. Abs. NLT 
90%) 
CLASS І 
e.g. Propranolol 
metoprolol 
CLASS II 
e.g. piroxicam, 
naproxen 
Low Permeability 
(Fract. Abs. 
<90%) 
CLASS III 
e.g. ranitidine 
cimetidine 
CLASS IV 
e.g. furosemide 
hydrochlorothiazide
Drug release mechanism in CDDS 
Slow zero order release 
Slow first order release 
Initial rapid release followed by slow zero order 
release 
Initial rapid release followed by slow first order 
release
• Zero order kinetics 
• First order kinetics 
• Hixoncrowell cube-root model 
• Higuchi model 
• Korsmeyer peppas model
• The equation for zero order kinetics is 
Qt=Qo+Kot 
Qo=initial amount of drug 
Qt=cumulative drug released at t 
Ko=zero order release constant 
t= time in hours 
• It describes the system where the drug release rate is 
independent of its concentration of the dissolved 
substance.
• A graph is plotted between the time taken on x axis 
and cumulative percentage of drug release on y axis 
and it gives straight line 
Time(hours) 
Cummulative %of drug release
• First order release equation is 
logQt=logQo + Kt/2.303 
Qt=cumulative amount of drug released at time t 
Qo= initial amount of drug 
K= first order rate constant 
t= time in hours 
• Here the drug release rate depends on concentration 
( unimolecular reaction)
• A graph is plotted between time taken on x- axis and 
log cumulative percentage of drug release on y- axis 
will give a straight line 
Time(hours) 
Log % of drug release
• THE HIXSON-CROWELL RELEASE EQUATION IS 
∛Qo-∛Qt=KHCt 
Qo=initial amount of drug 
Qt= cumulative amount of drug released at time t 
KHC= hixson crowell release constant 
t= time in hours 
• It describes the drug releases by dissolution and 
with the change in surface area and diameter of 
particle.
• A linear plot of cube root initial concentration minus 
cube root of percent remaining vs time gives straight 
line . 
• And release is dissolution rate controlled. 
• slope gives K value 
Time (hours) 
∛Qo-∛Qt
• Higuchi equation is 
Q=[Dt/Ƭ (2A-tCs)Cst]1/2 
or Q=Kt1/2 
Q=cumulative drug release at time t 
D= diffusion coefficient of drug in in matrix 
Cs=solubility of drug in polymeric matrix 
Ƭ= tortuosity of capillary system 
A=total amount of drug in unit volume matrix 
K= higuchi release constant 
• Higuchi equation suggest s that the drug release is by diffusion
• A graph is plotted with square root of time taken on x-axis 
and cumulative % of drug release on y- axis 
• It gives a straight line, slope gives K value 
√Time ( hours) 
cumulative % of drug release
• Korsmeyer –peppas equation is 
F=Mt/M∞ = Ktn 
F= fraction of drug released at time t 
Mt=amount of drug released at time t 
M∞=amount of drug released at infinite time 
K= kinetic constant 
t= time in hours 
n= diffusion or release exponent 
• n is estimated from linear regression of (Mt/M∞) vs time(t) 
• If n= 0.45 then indicate fickian diffusion 
• If o.45<n<0.89 then indicates anomalous diffusion or non-fickian 
diffusion 
• If n= 0.89 then indicates case-2relaxtaion or super case transport-2
• A graph is plotted between log time taken on x-axis 
and log of cumulative percentage of drug release at y-axis 
it gives a straight line. 
Log time 
cumulative % of drug release
Classification 
• There are basically three general 
categories of dissolution apparatus : 
1. Beaker methods 
2. Open flow-through compartment system 
3. Dialysis concept
1. BEAKER METHODS
to accessing therapeutic efficacy. 
Monitoring batch to batch consistency. 
High cost of in vitro dissolution test. 
Assessment of bioequivalence. 
Requirement for regulatory approval for 
product marketing and is a vital 
component of the quality control program. 
40
II..PP.. UUSSPP BB..PP.. EE..PP.. 
TTyyppee 11 
PPaaddddllee 
aappppaarraattuuss BBaasskkeett 
aappppaarraattuuss 
BBaasskkeett 
aappppaarraattuuss 
PPaaddddllee 
aappppaarraattuuss 
TTyyppee 22 BBaasskkeett 
aappppaarraattuuss 
PPaaddddllee 
aappppaarraattuuss 
PPaaddddllee 
aappppaarraattuuss 
BBaasskkeett 
aappppaarraattuuss 
TTyyppee 33 RReecciipprrooccaattiinngg 
ccyylliinnddeerr 
FFllooww tthhrroouugghh cceellll 
aappppaarraattuuss 
FFllooww tthhrroouugghh 
cceellll aappppaarraattuuss 
TTyyppee 44 FFllooww tthhrroouugghh 
cceellll aappppaarraattuuss 
TTyyppee 55 PPaaddddllee oovveerr 
ddiisskk 
TTyyppee 66 ccyylliinnddeerr 
TTyyppee 77 RReecciipprrooccaattiinngg 
hhoollddeerr 
41
Design: 
Vessel:- 
 Made up of borosilicate glass 
 semi hemispherical bottom 
 Capacity: 1000ml 
Shaft:- 
 Stainless steel 316 
 Rotates smoothly without significance wooble 
Basket:- 
 Stainless steel 316 
 Gold coatings up to 0.0001 inch 
Water bath:- Maintained at 37± 0.5˚c 
42
• Dosage form contained 
within basket 
• Dissolution should occur 
within Basket 
• pH change by media 
exchange 
• Uses: Capsules, 
tablets, delayed 
release, suppositories, 
floating dosage forms. 
43
• • 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 
44
45
• Dosage form should 
remain at the bottom centre 
of the vessel 
• Sinkers used for floaters 
• Useful for : 
• – Tablets 
• – Capsules 
• pH change by media 
addition 
46
• • Drug product 
– Solids (mostly non floating) 
• Monodisperse (tablets) 
• Polydisperse (encapsulated beads) 
• • Agitation 
– Rotating stirrer 
– Usual speed: 25 to 100 rpm 
• Standard volume: 900/1000 ml 
• Advantages: 
1. Easy to use and robust 
2. Ph change possible 
3. Can be easily adapted to apparatus 5 
• Disadvantages 
– Floating dosage forms require sinker 
– Positioning of tablet 
47
48
Design: 
1.vessel: cylindrical flat bottom glass vessel. 
2.Agitation type: -reciprocating 
-generally 5-35 rpm 
3. Volume of dissolution fluids: 200-250 ml 
4. Water bath: maintain at 37±0.5˚c 
5. Use: extended release 
49
The assembly consists of a 
set of cylindrical, flat-bottomed 
glass vessels; a 
set of glass reciprocating 
cylinders; )stainless steel 
fittings (type 316 or 
equivalent) and screens that 
are made of suitable 
nonsorbing and nonreactive 
aterial(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 
50
• 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 
51
Apparatus 3 – Reciprocating cylinder 
52
53
USP APPARATUS 4 - FLOW THROUGH 
CELL 
54
• 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 
55
Tablets 12 mm Tablets 22,6 mm Powders / Granules Implants Suppositories / 
Soft gelatine capsules 
• 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 
56
APPARATUS 4 – FLOW-THROUGH 
CELL 
57
Design 
1. Vessel: 
2. Shaft: 
3. Stirring elements 
4. Sample holder: - Disk assembly that hold the 
product in such a way that release surface is 
parallel with paddle. 
5. Paddle is directly attached over disk assembly. 
6. Samples are drawn away b/w the surface of 
medium and top of paddle blade. 
7. Volume; 900ml 
8. Temperature; 32˚c 
58
USP APPARATUS 5 - PADDLE OVER 
DISK 
59
• 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 60
Design; 
1. Vessel: in place of basket cylinder is used. 
2. Cylinder : stainless steel 316. 
3.Sample: - mounted to cuprophan(inner porous 
cellulosic material) an entire system is adhere to 
cylinder. 
-Dosage unit is place in cylinder and 
released from outside. 
4. Water bath : maintain at 32±.0.5˚c 
Use : transdermal patches can not be cut into small 
size. 
61
USP APPARATUS 6 - CYLINDER 
62
• 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. 
63
64
USP APPARATUS 7 – RECIPROCATING HOLDER 
65
• The assembly consists of a set of volumetrically 
calibrated solution containers made of glass 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 66
•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. 
67
Advantages of the Beaker 
Methods 
The basket method is the most widely used 
procedure which confines the solid dosage 
form to a limited area which is essential for 
better reproducibility. 
It is advantageous for capsules as they tend 
to float at the surface thus minimizing the 
area exposed to the dissolution fluid.
Limitation of the Beaker Methods 
Clogging of the basket screen by gummy particles. 
Tendency of the light particles to float. 
Sensitivity of the apparatus to variables such as 
vibration, eccentricity, etc. 
Rapid corrosion of the SS mesh in presence of 
HCl. 
Sensitivity of the apparatus to any slight changes 
in the paddle orientation. 
Non-reproducible position of the tablets at the 
bottom of the flask.
2. OPEN FLOW-THROUGH 
COMPARTMENT SYSTEM 
The dosage form is contained in a small vertical 
glass column with built in filter through which a 
continuous flow of the dissolution medium is 
circulated upward at a specific rate from an 
outside reservoir using a peristaltic or centrifugal 
pump. 
Dissolution fluid is collected in a separate 
reservoir. 
E.g. lipid filled soft Gelatin capsule
Advantages 
No stirring and drug particles are exposed 
to homogeneous, laminar flow that can be 
precisely controlled. All the problems of 
wobbling, shaft eccentricity, vibration, 
stirrer position don’t exist. 
There is no physical abrasion of solids. 
Perfect sink conditions can be maintained.
Disadvantages 
Tendency of the filter to clog because of 
the unidirectional flow. 
Different types of pumps, such as 
peristaltic and centrifugal, have been 
shown to give different dissolution results. 
Temperature control is also much more 
difficult to achieve in column type flow 
through system than in the conventional 
stirred vessel type.
3. DIALYSIS SYSTEM 
Here, dialysis membrane used as a 
selective barrier between fresh solvent 
compartment and the cell compartment 
containing dosage form. 
It can be used in case of very poorly 
soluble rugs and dosage form such as 
ointments, creams and suspensions.
THE ROTATING FILTER METHOD 
It consists of a magnetically driven rotating 
filter assembly and a 12 mesh wire cloth 
basket. 
The sample is withdrawn through the 
spinning filter for analysis.
ROTATING FLASK DISSOLUTION 
METHOD 
This consists of a spherical flask made of 
glass and supported by a horizontal glass 
shaft that is fused to its sides. 
The shaft is connected to a constant 
speed driving motor. 
The flask is placed in a constant 
temperature water bath and rotates about 
its horizontal axis.
ROTATING AND STATIC DISK 
METHODS 
The compound is 
compressed into non 
disintegrating disc 
Mounted – One surface 
is exposed to medium 
Assumption – Surface 
area remains constant 
Used to determine the 
intrinsic dissolution rate
Types of dosage form Release method 
Solid oral dosage forms(conventional) Basket,paddle,reciprocating cylinder, 
or flow through cell 
Oral suspension Paddle 
Orally disintegrating tablets Paddle 
Chewable tablets Basket,paddle,reciprocating cylinder 
Transdermal-patches Paddle over disk 
Suppositories Paddle, modified basket, or dual 
chamber flow through cell 
80
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 
81
FACTORS RELATED TO PHYSICOCHEMICAL 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 :neomycin 
82
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. 83
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. 84
• 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 
85
• 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. 
86
• 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 
87
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 or 
Formation 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 
88
FACTORS RELATED TO DRUG 
PRODUCT 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 
89 
FACTORS RELATED TO DRUG 
PRODUCT FORMULATION
10 20 30 40 50 
Time in min. 
100 
80 
60 
40 
20 
Amt of dissolved mg 
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). 
90
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. 
91
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. 
92
• 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% gelatin whereas decrease in dissolution rate with 20% gelatin. 
This was due to higher concentration which formed a thick film 
around tablet. 
• Water soluble granulating agent Plasdone gives faster dissolution 
rate compared to gelatin. 
93
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. 
94
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. 
95
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 
96
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. 
97
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. 
98
2)COMPRESSION FORCE 
• The compression process influence density, porosity, hardness, 
disintegration time & dissolution of tablet. 
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 
99
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 
• Similarly as increase in mixing time of formulation 
containing 97 to 99% microcrystalline cellulose or 
another slightly swelling disintegrant result in enhance 
dissolution rate. 
100
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. 
101
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. 
102
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%. 
103
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. 
• Accumulation of the particulate matter on the surface 
may cause significant error in the dissolution testing. 
104
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 
105
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 leads to floating of powder which leads to 
wetting and penetration problem. 
• Dissolution media composition & PH 
 Addition of Na – sulfate decrease the dissolution rate. 
 Addition of urea increase dissolution rate. 
106
• 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. 
107
• In order to minimize the effect of conc. gradient and maintain sink 
conditions, the conc. of drug should not exceed 10-15% of its maximum 
Solubility in dissolution medium selected. 
• However, some insoluble drug a 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 
dissolution 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%). 
108
DISSOLUTION MEDIUM EXAMPLE 
Water Ampicillin caps., butabarbital 
sodium tabs. 
Buffers Azithromycin caps., 
paracetamol tabs. 
HCL solution Cemetidine tabs. 
Simulated gastric fluid Astemizole tabs., piroxicam 
caps. 
Simulated intestinal fluid Valproic caps., Glipizide tabs. 
Surfactant solution Clofibrate caps, danazol caps
DISSOLUTION ACCEPTANCE 
CRITRIA 
• Q –Value – 
• Define as a percentage of drug conten 
dissolved in a given time period.
DISSOLUTION ACCEPTANCE CRITRIA 
STAGE No. of Dosage units 
tested 
Acceptance criteria 
S1 6 No Dosage unit is 
less then Q+5% 
S2 6 Average of 12 
dosage units (S1+S2) 
and no dosage unit is 
less then Q-15% 
S3 12(6+6+12=24) Average of 24 
dosage units >- And 
not more than two 
dosage units are less 
than Q-15% and No 
dosage unit is less 
than Q-25%
Method for comparison of 
dissolution profile 
• Difference factor (F1 Value)- 
• Define as calculate the % Difference 
between 2 curves at each time point and 
is a measurement of the relative error 
between 2 curves. 
• f1= {[Σ t=1n |Rt-Tt|] / [Σ t=1n Rt]} ×100. 
• Values range from 0 to 15
• Similarity Factor (F 2 value)-define as 
measurement of similarity in % Dissolution 
between two curve. 
• Where Rt and Tt = cumulative % dissolved 
for reference and test 
• Values range from 50 to 100 113
USP SGF (simulated gastric fluid) 
NaCl 2.0 g 
Purified pepsin 3.2 g 
HCl 7.0 mL 
Purified water qs. 1000 mL 
Media has a pH of about 1.2 
USP SIF (simulated intestinal fluid) 
Monobasic potassium phosphate 6.8 g in Purified 
water 250 mL 
NaOH (0.2 N) 77 mL and Purified water 500 mL 
Pancreatin 10.0 g 
Adjust with either 0.2 N NaOH or 0.2 N HCl to a pH 
of 6.8 ± 0.1. 
Purified water qs. 1000 mL
Oral Drug Absorption 
Gastric 
Emptying 
Transit Permeation 
Dissolution 
Metabolism
In Vivo and In Vitro Relationship: 
Scientific Issues 
• Limits to oral drug 
absorption 
– Dissolution-limited 
– Solubility-limited 
– Permeability-limited 
Gastric 
Emptying 
Transit Permeation 
Dissolution 
Metabolism 
SSoolluubbiilliittyy 
Dissolution Permeation 
Conc ³ Solubility
IN VITRO IN VIVO 
CORRELATION
In vitro-in vivo correlation 
• A predictive mathematical model 
that describes the relationship 
between an in-vitro property of a 
dosage form and an in-vivo 
response.
• Key goal in development of dosage form 
and good understanding of in vitro and in 
vivo performance of dosage form 
• Formulation optimization requires altering 
some parameters – bioavailability studies 
• Regulatory guidance developed to 
minimize the additional bioavailability 
studies
120 
Purpose of IVIVC 
• The optimization of formulations 
– may require changes in the composition, 
manufacturing process, equipment, and batch sizes. 
– In order to prove the validity of a new formulation, 
which is bioequivalent with a target formulation, a 
considerable amount of efforts is required to study 
bioequivalence (BE)/bioavailability(BA). 
• The main purpose of an IVIVC model 
– to utilize in vitro dissolution profiles as a surrogate for 
in vivo bioequivalence and to support biowaivers 
– Data analysis of IVIVC attracts attention from the 
pharmaceutical industry.
Basic approaches 
• By establishing a relationship usually 
linear, between the in vitro dissolution and 
in vivo bioavailability parameters. 
• By using data from previous bioavailability 
studies to modify the dissolution 
methodology.
• In vitro – in vivo correlation can be achieved 
using 
 Pharmacological correlation 
 Semi quantitative correlation 
 Quantitative correlation
In vitro-in vivo correlations 
• Correlations based on the plasma level 
data 
• Correlations based on the urinary 
excretion data 
• Correlations based on the 
pharmacological response
DEFINITION 
• USP definition 
“The establishment of rational relationship b/w a biological 
property or a parameter derived from a biological 
property produced by a dosage form and 
physicochemical property of same dosage form” 
• FDA definition 
“It is predictive mathematical model describing the 
relationship b/w in vitro property of dosage form and a 
relevant in vivo response”
IMPORTANCE 
• Serves as a surrogate of in vivo and assist in 
supporting biowaivers 
• Validates the use of dissolution methods and 
specification 
• Assist in QC during mfg and selecting the 
appropriate formulation
IVIVC levels 
• Level A: 
– Point to point correlation is developed between in vitro 
dissolution rate and the in vivo rate of absorption 
• Level B: 
– Utilises statistical moment analysis and the mean in vitro 
dissolution time is compared to either the mean residence time 
or the mean in vivo dissolution time 
• Level C: 
– single point correlation that relates one dissolution time point to 
one pharmacokinetic parameter 
Multiple level C
Level A correlation 
• Highest category 
correlation 
• Represents point to 
point relationship 
• Developed by two 
stage procedure 
Deconvulation 
Comparison 
• Purpose – define 
direct relationship 
120 
100 
80 
60 
40 
20 
0 
0 20 40 60 80 100 120 
% Drug Dissolved 
% Drug 
Absorbed
Level B correlation 
• Utilizes the principle of statistical moment 
analysis 
MDT vitro is compared with MRT vivo 
• No point to point correlation 
• Does not reflect the actual in vivo plasma level 
curves
Level C correlation 
• Dissolution time point (t 50%,t 90% ) is 
compared to one mean pharmacokinetic 
parameter ( Cmax ,tmax ,AUC) 
• Single point correlation 
• Weakest level of correlation as partial 
relationship b/w absorption and dissolution 
is established 
• Useful in the early stages of formulation 
development
Multiple level C correlation 
• It reflects the relationship b/w one or several 
pharmacokinetic parameter of interest and 
amount of drug dissolved at several time point of 
dissolution profile 
• Base on 
 Early 
 Middle 
 Late stage
Applications 
• Ensure batch to batch consistency 
• Serve as a tool in the development of a 
new dosage form with desired in-vivo 
performance 
• Assist in validating or setting dissolution 
specifications

More Related Content

What's hot

Consolidation, effect of friction, distribution of forces, compaction profile
Consolidation, effect of friction, distribution of forces, compaction profileConsolidation, effect of friction, distribution of forces, compaction profile
Consolidation, effect of friction, distribution of forces, compaction profile
Zahid1392
 

What's hot (20)

Dissolution models (sem 1)
Dissolution models (sem 1)Dissolution models (sem 1)
Dissolution models (sem 1)
 
Problems of variable
Problems of variableProblems of variable
Problems of variable
 
Dissolution method and ivivc by ranjeet singh
Dissolution method and ivivc by ranjeet singhDissolution method and ivivc by ranjeet singh
Dissolution method and ivivc by ranjeet singh
 
Modified drug delivery systems, Targeted drug delivery and biopharmaceutical ...
Modified drug delivery systems, Targeted drug delivery and biopharmaceutical ...Modified drug delivery systems, Targeted drug delivery and biopharmaceutical ...
Modified drug delivery systems, Targeted drug delivery and biopharmaceutical ...
 
generic substitution and biowaver
generic substitution and biowavergeneric substitution and biowaver
generic substitution and biowaver
 
Physics of tablet compression
Physics of tablet compressionPhysics of tablet compression
Physics of tablet compression
 
Consolidation, effect of friction, distribution of forces, compaction profile
Consolidation, effect of friction, distribution of forces, compaction profileConsolidation, effect of friction, distribution of forces, compaction profile
Consolidation, effect of friction, distribution of forces, compaction profile
 
Drug excipient interaction different method
Drug excipient interaction different methodDrug excipient interaction different method
Drug excipient interaction different method
 
Techniques for enhancement of dissolution rate
Techniques for enhancement of dissolution rateTechniques for enhancement of dissolution rate
Techniques for enhancement of dissolution rate
 
Drug excipient interaction
Drug excipient interaction Drug excipient interaction
Drug excipient interaction
 
Biological process involved in drug targetting
Biological process involved  in drug targettingBiological process involved  in drug targetting
Biological process involved in drug targetting
 
Biopharmaceutic considerations in drug product design and In Vitro Drug Produ...
Biopharmaceutic considerations in drug product design and In Vitro Drug Produ...Biopharmaceutic considerations in drug product design and In Vitro Drug Produ...
Biopharmaceutic considerations in drug product design and In Vitro Drug Produ...
 
THE FACTOR AFFECTING DISSOLUTION RATE
THE FACTOR AFFECTING DISSOLUTION RATE THE FACTOR AFFECTING DISSOLUTION RATE
THE FACTOR AFFECTING DISSOLUTION RATE
 
Physics of tablet compression
Physics of tablet compressionPhysics of tablet compression
Physics of tablet compression
 
permeability in vivo in vitro in situ.pptx
permeability in vivo in vitro in situ.pptxpermeability in vivo in vitro in situ.pptx
permeability in vivo in vitro in situ.pptx
 
Compendial methods of dissolution testing
Compendial methods of dissolution testing Compendial methods of dissolution testing
Compendial methods of dissolution testing
 
Drug product performance
Drug product performanceDrug product performance
Drug product performance
 
Rate limiting steps in drug absorption
Rate limiting steps in drug absorptionRate limiting steps in drug absorption
Rate limiting steps in drug absorption
 
Modified release drug products
Modified release drug productsModified release drug products
Modified release drug products
 
Drug product performance , in vivo: bioavailability and bioequivalence
Drug product performance , in vivo: bioavailability and bioequivalenceDrug product performance , in vivo: bioavailability and bioequivalence
Drug product performance , in vivo: bioavailability and bioequivalence
 

Viewers also liked

Dissolution test.
Dissolution test.Dissolution test.
Dissolution test.
Salum Mkata
 
Dissolution testing
Dissolution testingDissolution testing
Dissolution testing
Gaurav Kr
 

Viewers also liked (20)

Dissolution
DissolutionDissolution
Dissolution
 
Dissolution
DissolutionDissolution
Dissolution
 
drug dissolution
drug dissolutiondrug dissolution
drug dissolution
 
Dissolution
DissolutionDissolution
Dissolution
 
Dissolution and In Vitro In Vivo Correlation (IVIVC)
Dissolution and In Vitro In Vivo Correlation (IVIVC)Dissolution and In Vitro In Vivo Correlation (IVIVC)
Dissolution and In Vitro In Vivo Correlation (IVIVC)
 
Theories of drug dissolution
Theories of drug dissolutionTheories of drug dissolution
Theories of drug dissolution
 
Theories of Dissolution
Theories of DissolutionTheories of Dissolution
Theories of Dissolution
 
Dissolution study-Dissolution studies Factor affecting dissolution and Invitr...
Dissolution study-Dissolution studies Factor affecting dissolution and Invitr...Dissolution study-Dissolution studies Factor affecting dissolution and Invitr...
Dissolution study-Dissolution studies Factor affecting dissolution and Invitr...
 
Dissolution
DissolutionDissolution
Dissolution
 
In vitro Dissolution Testing Models
In vitro Dissolution Testing ModelsIn vitro Dissolution Testing Models
In vitro Dissolution Testing Models
 
Protein drug binding
Protein drug bindingProtein drug binding
Protein drug binding
 
Protein Binding of Drugs by Dr. Sanaullah Aslam
Protein Binding of Drugs by Dr. Sanaullah AslamProtein Binding of Drugs by Dr. Sanaullah Aslam
Protein Binding of Drugs by Dr. Sanaullah Aslam
 
Theories of dissolution
Theories of dissolutionTheories of dissolution
Theories of dissolution
 
Dissolution test.
Dissolution test.Dissolution test.
Dissolution test.
 
Drug release kinetics
Drug release kineticsDrug release kinetics
Drug release kinetics
 
Dissolution testing
Dissolution testingDissolution testing
Dissolution testing
 
DISSOLUTION TESTING APPARATUS
DISSOLUTION TESTING APPARATUSDISSOLUTION TESTING APPARATUS
DISSOLUTION TESTING APPARATUS
 
Pharmacokinetic models
Pharmacokinetic modelsPharmacokinetic models
Pharmacokinetic models
 
Sustained release dosage form
Sustained release dosage formSustained release dosage form
Sustained release dosage form
 
Drug release and dissolution
Drug release and dissolution Drug release and dissolution
Drug release and dissolution
 

Similar to M pharm dissolution

Dissolution study
Dissolution  studyDissolution  study
Dissolution study
CPATRO
 
10. Mass Transport andert Dissolution.pptx
10. Mass Transport andert Dissolution.pptx10. Mass Transport andert Dissolution.pptx
10. Mass Transport andert Dissolution.pptx
interaman123
 

Similar to M pharm dissolution (20)

Theory of Dissolution.pdf
Theory of Dissolution.pdfTheory of Dissolution.pdf
Theory of Dissolution.pdf
 
Kausar
KausarKausar
Kausar
 
Kausar
KausarKausar
Kausar
 
Dissolution study
Dissolution  studyDissolution  study
Dissolution study
 
DISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUSDISSOLUTION PARAMETERS AND ITS APPARATUS
DISSOLUTION PARAMETERS AND ITS APPARATUS
 
Different theory of drug dissolution
Different theory of drug dissolutionDifferent theory of drug dissolution
Different theory of drug dissolution
 
Theories
TheoriesTheories
Theories
 
disso models ppt
 disso models ppt disso models ppt
disso models ppt
 
Theories of dissolution
Theories of dissolutionTheories of dissolution
Theories of dissolution
 
10. Mass Transport andert Dissolution.pptx
10. Mass Transport andert Dissolution.pptx10. Mass Transport andert Dissolution.pptx
10. Mass Transport andert Dissolution.pptx
 
Consolidation parameter
Consolidation parameterConsolidation parameter
Consolidation parameter
 
Consolidation parameter
Consolidation parameterConsolidation parameter
Consolidation parameter
 
Factors influencing gi absorption of a drug new
Factors influencing gi absorption of a drug  newFactors influencing gi absorption of a drug  new
Factors influencing gi absorption of a drug new
 
Consolidation parameters
Consolidation parametersConsolidation parameters
Consolidation parameters
 
Dissolution final
Dissolution final Dissolution final
Dissolution final
 
Dissolution kinetics and dissolutition modesl
Dissolution kinetics and dissolutition modeslDissolution kinetics and dissolutition modesl
Dissolution kinetics and dissolutition modesl
 
Bhavani sem
Bhavani semBhavani sem
Bhavani sem
 
DISSOLUTION AND MECHANISM OF DRUG RELEASE
 DISSOLUTION AND MECHANISM OF DRUG RELEASE DISSOLUTION AND MECHANISM OF DRUG RELEASE
DISSOLUTION AND MECHANISM OF DRUG RELEASE
 
Dissolution v2
Dissolution v2Dissolution v2
Dissolution v2
 
Diffusion (Physical Pharmacy)
Diffusion (Physical Pharmacy)Diffusion (Physical Pharmacy)
Diffusion (Physical Pharmacy)
 

More from Malla Reddy College of Pharmacy

More from Malla Reddy College of Pharmacy (20)

Rna secondary structure prediction
Rna secondary structure predictionRna secondary structure prediction
Rna secondary structure prediction
 
Proteomics
ProteomicsProteomics
Proteomics
 
Proteins basics
Proteins basicsProteins basics
Proteins basics
 
Protein structure classification
Protein structure classificationProtein structure classification
Protein structure classification
 
Protein identication characterization
Protein identication characterizationProtein identication characterization
Protein identication characterization
 
Protein modeling
Protein modelingProtein modeling
Protein modeling
 
Primerdesign
PrimerdesignPrimerdesign
Primerdesign
 
Phylogenetic studies
Phylogenetic studiesPhylogenetic studies
Phylogenetic studies
 
Multiple sequence alignment
Multiple sequence alignmentMultiple sequence alignment
Multiple sequence alignment
 
Homology modeling tools
Homology modeling toolsHomology modeling tools
Homology modeling tools
 
Homology modeling
Homology modelingHomology modeling
Homology modeling
 
Genome assembly
Genome assemblyGenome assembly
Genome assembly
 
Genome analysis2
Genome analysis2Genome analysis2
Genome analysis2
 
Genome analysis
Genome analysisGenome analysis
Genome analysis
 
Fasta
FastaFasta
Fasta
 
Drug design intro
Drug design introDrug design intro
Drug design intro
 
Drug design
Drug designDrug design
Drug design
 
Data retrieval
Data retrievalData retrieval
Data retrieval
 
Blast
BlastBlast
Blast
 
Biological databases
Biological databasesBiological databases
Biological databases
 

M pharm dissolution

  • 2. Definition- • Dissolution is a process in which a solid substance solubilizes in a given solvent i.e. mass transfer from the solid surface to the liquid phase. • Rate of dissolution is the amount of drug substance that goes in solution per unit time under standardized conditions of liquid/solid interface, temperature and solvent composition. 2
  • 4. • Initial mechanical lag • Wetting of dosage form • Penetration of dissolution medium • Disintegration • Deaggregation • Dissolution • Occlusion of some particles 4
  • 5.  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. 5
  • 6. 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 - DDiiffffuussiioonn ccooeeffffiicciieenntt, 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 6
  • 8. Theories of Drug Dissolution 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.
  • 9. I. Diffusion layer model/Film Theory :- • It involves two steps :- a. Solution of the solid to form stagnant film or diffusive layer which is saturated with the drug b. Diffusion of the soluble solute from the stagnant layer to the bulk of the solution; this is r.d.s in drug dissolution.
  • 10.
  • 11. • The rate of dissolution is given by Noyes and Whitney: = k (Cs- Cb) dc dt Where, dc/dt= dissolution rate of the drug K= dissolution rate constant Cs= concentration of drug in stagnant layer Cb= concentration of drug in the bulk of the solution at time t
  • 12. Modified Noyes-Whitney’s Equation - 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.
  • 13. Sink condition A 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. 13
  • 14. • Dissolution rate under sink condition follow zero order dissolution rate. Conc of disslove drug Time Zero order dissolution Under sink condition First order under non sink condition 14
  • 15. 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. 15
  • 16.  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 16
  • 17. • This is first order dissolution rate process, for which the driving force is concentration gradient. • This is true for in-vitro dissolution which is characterized by non-sink conditions. • The in-vivo dissolution is rapid as sink conditions are maintained by absorption of drug in systemic circulation i.e. Cb=0 and rate of dissolution is maximum.
  • 18. • Under sink conditions, if the volume and surface area of the solid are kept constant, then dC dt = K • This represents that the dissolution rate is constant under sink conditions and follows zero order kinetics.
  • 19. II. Danckwert’s model/Penetration or surface renewal Theory :- • Dankwert takes into account the eddies or packets that are present in the agitated fluid which reach the solid-liquid interface, absorb the solute by diffusion and carry it into the bulk of solution. • These packets get continuously replaced by new ones and expose to new solid surface each time, thus the theory is called as surface renewal theory.
  • 20.
  • 21. • The Danckwert’s model is expressed by equation dC dt = dm V = A (Cs-Cb). γ D dt Where, m = mass of solid dissolved Gamma (γ) = rate of surface renewal
  • 22. III. Interfacial barrier model/Double barrier or Limited solvation theory :- • The concept of this theory is explained by following equation- G = Ki (Cs - Cb) Where, G = dissolution rate per unit area, Ki = effective interfacial transport constant.
  • 23. 3) Interfacial layer model S Film boundary Bulk solution Cs C Stagnant layer In this model it is assumed that the reaction at solid surface is not instantaneous i.e. the reaction at solid surface and its diffusion across the interface is slower than diffusion across liquid film. therefore the rate of solubility of solid in liquid film becomes the rate limiting than the diffusion of dissolved molecules equation : dm/dt = Ki (Cs – C ) K = effective interfacial transport rate constant
  • 24. Biopharmaceutical Classification System High Solubility (Dose Vol. NMT 250 mL) Low Solubility (Dose Vol. >250 mL) High Permeability (Fract. Abs. NLT 90%) CLASS І e.g. Propranolol metoprolol CLASS II e.g. piroxicam, naproxen Low Permeability (Fract. Abs. <90%) CLASS III e.g. ranitidine cimetidine CLASS IV e.g. furosemide hydrochlorothiazide
  • 25. Drug release mechanism in CDDS Slow zero order release Slow first order release Initial rapid release followed by slow zero order release Initial rapid release followed by slow first order release
  • 26. • Zero order kinetics • First order kinetics • Hixoncrowell cube-root model • Higuchi model • Korsmeyer peppas model
  • 27. • The equation for zero order kinetics is Qt=Qo+Kot Qo=initial amount of drug Qt=cumulative drug released at t Ko=zero order release constant t= time in hours • It describes the system where the drug release rate is independent of its concentration of the dissolved substance.
  • 28. • A graph is plotted between the time taken on x axis and cumulative percentage of drug release on y axis and it gives straight line Time(hours) Cummulative %of drug release
  • 29. • First order release equation is logQt=logQo + Kt/2.303 Qt=cumulative amount of drug released at time t Qo= initial amount of drug K= first order rate constant t= time in hours • Here the drug release rate depends on concentration ( unimolecular reaction)
  • 30. • A graph is plotted between time taken on x- axis and log cumulative percentage of drug release on y- axis will give a straight line Time(hours) Log % of drug release
  • 31. • THE HIXSON-CROWELL RELEASE EQUATION IS ∛Qo-∛Qt=KHCt Qo=initial amount of drug Qt= cumulative amount of drug released at time t KHC= hixson crowell release constant t= time in hours • It describes the drug releases by dissolution and with the change in surface area and diameter of particle.
  • 32. • A linear plot of cube root initial concentration minus cube root of percent remaining vs time gives straight line . • And release is dissolution rate controlled. • slope gives K value Time (hours) ∛Qo-∛Qt
  • 33. • Higuchi equation is Q=[Dt/Ƭ (2A-tCs)Cst]1/2 or Q=Kt1/2 Q=cumulative drug release at time t D= diffusion coefficient of drug in in matrix Cs=solubility of drug in polymeric matrix Ƭ= tortuosity of capillary system A=total amount of drug in unit volume matrix K= higuchi release constant • Higuchi equation suggest s that the drug release is by diffusion
  • 34. • A graph is plotted with square root of time taken on x-axis and cumulative % of drug release on y- axis • It gives a straight line, slope gives K value √Time ( hours) cumulative % of drug release
  • 35. • Korsmeyer –peppas equation is F=Mt/M∞ = Ktn F= fraction of drug released at time t Mt=amount of drug released at time t M∞=amount of drug released at infinite time K= kinetic constant t= time in hours n= diffusion or release exponent • n is estimated from linear regression of (Mt/M∞) vs time(t) • If n= 0.45 then indicate fickian diffusion • If o.45<n<0.89 then indicates anomalous diffusion or non-fickian diffusion • If n= 0.89 then indicates case-2relaxtaion or super case transport-2
  • 36. • A graph is plotted between log time taken on x-axis and log of cumulative percentage of drug release at y-axis it gives a straight line. Log time cumulative % of drug release
  • 37.
  • 38. Classification • There are basically three general categories of dissolution apparatus : 1. Beaker methods 2. Open flow-through compartment system 3. Dialysis concept
  • 40. to accessing therapeutic efficacy. Monitoring batch to batch consistency. High cost of in vitro dissolution test. Assessment of bioequivalence. Requirement for regulatory approval for product marketing and is a vital component of the quality control program. 40
  • 41. II..PP.. UUSSPP BB..PP.. EE..PP.. TTyyppee 11 PPaaddddllee aappppaarraattuuss BBaasskkeett aappppaarraattuuss BBaasskkeett aappppaarraattuuss PPaaddddllee aappppaarraattuuss TTyyppee 22 BBaasskkeett aappppaarraattuuss PPaaddddllee aappppaarraattuuss PPaaddddllee aappppaarraattuuss BBaasskkeett aappppaarraattuuss TTyyppee 33 RReecciipprrooccaattiinngg ccyylliinnddeerr FFllooww tthhrroouugghh cceellll aappppaarraattuuss FFllooww tthhrroouugghh cceellll aappppaarraattuuss TTyyppee 44 FFllooww tthhrroouugghh cceellll aappppaarraattuuss TTyyppee 55 PPaaddddllee oovveerr ddiisskk TTyyppee 66 ccyylliinnddeerr TTyyppee 77 RReecciipprrooccaattiinngg hhoollddeerr 41
  • 42. Design: Vessel:-  Made up of borosilicate glass  semi hemispherical bottom  Capacity: 1000ml Shaft:-  Stainless steel 316  Rotates smoothly without significance wooble Basket:-  Stainless steel 316  Gold coatings up to 0.0001 inch Water bath:- Maintained at 37± 0.5˚c 42
  • 43. • Dosage form contained within basket • Dissolution should occur within Basket • pH change by media exchange • Uses: Capsules, tablets, delayed release, suppositories, floating dosage forms. 43
  • 44. • • 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 44
  • 45. 45
  • 46. • Dosage form should remain at the bottom centre of the vessel • Sinkers used for floaters • Useful for : • – Tablets • – Capsules • pH change by media addition 46
  • 47. • • Drug product – Solids (mostly non floating) • Monodisperse (tablets) • Polydisperse (encapsulated beads) • • Agitation – Rotating stirrer – Usual speed: 25 to 100 rpm • Standard volume: 900/1000 ml • Advantages: 1. Easy to use and robust 2. Ph change possible 3. Can be easily adapted to apparatus 5 • Disadvantages – Floating dosage forms require sinker – Positioning of tablet 47
  • 48. 48
  • 49. Design: 1.vessel: cylindrical flat bottom glass vessel. 2.Agitation type: -reciprocating -generally 5-35 rpm 3. Volume of dissolution fluids: 200-250 ml 4. Water bath: maintain at 37±0.5˚c 5. Use: extended release 49
  • 50. The assembly consists of a set of cylindrical, flat-bottomed glass vessels; a set of glass reciprocating cylinders; )stainless steel fittings (type 316 or equivalent) and screens that are made of suitable nonsorbing and nonreactive aterial(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 50
  • 51. • 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 51
  • 52. Apparatus 3 – Reciprocating cylinder 52
  • 53. 53
  • 54. USP APPARATUS 4 - FLOW THROUGH CELL 54
  • 55. • 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 55
  • 56. Tablets 12 mm Tablets 22,6 mm Powders / Granules Implants Suppositories / Soft gelatine capsules • 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 56
  • 57. APPARATUS 4 – FLOW-THROUGH CELL 57
  • 58. Design 1. Vessel: 2. Shaft: 3. Stirring elements 4. Sample holder: - Disk assembly that hold the product in such a way that release surface is parallel with paddle. 5. Paddle is directly attached over disk assembly. 6. Samples are drawn away b/w the surface of medium and top of paddle blade. 7. Volume; 900ml 8. Temperature; 32˚c 58
  • 59. USP APPARATUS 5 - PADDLE OVER DISK 59
  • 60. • 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 60
  • 61. Design; 1. Vessel: in place of basket cylinder is used. 2. Cylinder : stainless steel 316. 3.Sample: - mounted to cuprophan(inner porous cellulosic material) an entire system is adhere to cylinder. -Dosage unit is place in cylinder and released from outside. 4. Water bath : maintain at 32±.0.5˚c Use : transdermal patches can not be cut into small size. 61
  • 62. USP APPARATUS 6 - CYLINDER 62
  • 63. • 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. 63
  • 64. 64
  • 65. USP APPARATUS 7 – RECIPROCATING HOLDER 65
  • 66. • The assembly consists of a set of volumetrically calibrated solution containers made of glass 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 66
  • 67. •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. 67
  • 68. Advantages of the Beaker Methods The basket method is the most widely used procedure which confines the solid dosage form to a limited area which is essential for better reproducibility. It is advantageous for capsules as they tend to float at the surface thus minimizing the area exposed to the dissolution fluid.
  • 69. Limitation of the Beaker Methods Clogging of the basket screen by gummy particles. Tendency of the light particles to float. Sensitivity of the apparatus to variables such as vibration, eccentricity, etc. Rapid corrosion of the SS mesh in presence of HCl. Sensitivity of the apparatus to any slight changes in the paddle orientation. Non-reproducible position of the tablets at the bottom of the flask.
  • 70. 2. OPEN FLOW-THROUGH COMPARTMENT SYSTEM The dosage form is contained in a small vertical glass column with built in filter through which a continuous flow of the dissolution medium is circulated upward at a specific rate from an outside reservoir using a peristaltic or centrifugal pump. Dissolution fluid is collected in a separate reservoir. E.g. lipid filled soft Gelatin capsule
  • 71.
  • 72. Advantages No stirring and drug particles are exposed to homogeneous, laminar flow that can be precisely controlled. All the problems of wobbling, shaft eccentricity, vibration, stirrer position don’t exist. There is no physical abrasion of solids. Perfect sink conditions can be maintained.
  • 73. Disadvantages Tendency of the filter to clog because of the unidirectional flow. Different types of pumps, such as peristaltic and centrifugal, have been shown to give different dissolution results. Temperature control is also much more difficult to achieve in column type flow through system than in the conventional stirred vessel type.
  • 74. 3. DIALYSIS SYSTEM Here, dialysis membrane used as a selective barrier between fresh solvent compartment and the cell compartment containing dosage form. It can be used in case of very poorly soluble rugs and dosage form such as ointments, creams and suspensions.
  • 75.
  • 76.
  • 77. THE ROTATING FILTER METHOD It consists of a magnetically driven rotating filter assembly and a 12 mesh wire cloth basket. The sample is withdrawn through the spinning filter for analysis.
  • 78. ROTATING FLASK DISSOLUTION METHOD This consists of a spherical flask made of glass and supported by a horizontal glass shaft that is fused to its sides. The shaft is connected to a constant speed driving motor. The flask is placed in a constant temperature water bath and rotates about its horizontal axis.
  • 79. ROTATING AND STATIC DISK METHODS The compound is compressed into non disintegrating disc Mounted – One surface is exposed to medium Assumption – Surface area remains constant Used to determine the intrinsic dissolution rate
  • 80. Types of dosage form Release method Solid oral dosage forms(conventional) Basket,paddle,reciprocating cylinder, or flow through cell Oral suspension Paddle Orally disintegrating tablets Paddle Chewable tablets Basket,paddle,reciprocating cylinder Transdermal-patches Paddle over disk Suppositories Paddle, modified basket, or dual chamber flow through cell 80
  • 81. 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 81
  • 82. FACTORS RELATED TO PHYSICOCHEMICAL 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 :neomycin 82
  • 83. 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. 83
  • 84. 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. 84
  • 85. • 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 85
  • 86. • 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. 86
  • 87. • 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 87
  • 88. 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 or Formation 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 88
  • 89. FACTORS RELATED TO DRUG PRODUCT 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 89 FACTORS RELATED TO DRUG PRODUCT FORMULATION
  • 90. 10 20 30 40 50 Time in min. 100 80 60 40 20 Amt of dissolved mg 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). 90
  • 91. 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. 91
  • 92. 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. 92
  • 93. • 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% gelatin whereas decrease in dissolution rate with 20% gelatin. This was due to higher concentration which formed a thick film around tablet. • Water soluble granulating agent Plasdone gives faster dissolution rate compared to gelatin. 93
  • 94. 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. 94
  • 95. 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. 95
  • 96. 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 96
  • 97. 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. 97
  • 98. 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. 98
  • 99. 2)COMPRESSION FORCE • The compression process influence density, porosity, hardness, disintegration time & dissolution of tablet. 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 99
  • 100. 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 • Similarly as increase in mixing time of formulation containing 97 to 99% microcrystalline cellulose or another slightly swelling disintegrant result in enhance dissolution rate. 100
  • 101. 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. 101
  • 102. 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. 102
  • 103. 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%. 103
  • 104. 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. • Accumulation of the particulate matter on the surface may cause significant error in the dissolution testing. 104
  • 105. 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 105
  • 106. 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 leads to floating of powder which leads to wetting and penetration problem. • Dissolution media composition & PH  Addition of Na – sulfate decrease the dissolution rate.  Addition of urea increase dissolution rate. 106
  • 107. • 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. 107
  • 108. • In order to minimize the effect of conc. gradient and maintain sink conditions, the conc. of drug should not exceed 10-15% of its maximum Solubility in dissolution medium selected. • However, some insoluble drug a 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 dissolution 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%). 108
  • 109. DISSOLUTION MEDIUM EXAMPLE Water Ampicillin caps., butabarbital sodium tabs. Buffers Azithromycin caps., paracetamol tabs. HCL solution Cemetidine tabs. Simulated gastric fluid Astemizole tabs., piroxicam caps. Simulated intestinal fluid Valproic caps., Glipizide tabs. Surfactant solution Clofibrate caps, danazol caps
  • 110. DISSOLUTION ACCEPTANCE CRITRIA • Q –Value – • Define as a percentage of drug conten dissolved in a given time period.
  • 111. DISSOLUTION ACCEPTANCE CRITRIA STAGE No. of Dosage units tested Acceptance criteria S1 6 No Dosage unit is less then Q+5% S2 6 Average of 12 dosage units (S1+S2) and no dosage unit is less then Q-15% S3 12(6+6+12=24) Average of 24 dosage units >- And not more than two dosage units are less than Q-15% and No dosage unit is less than Q-25%
  • 112. Method for comparison of dissolution profile • Difference factor (F1 Value)- • Define as calculate the % Difference between 2 curves at each time point and is a measurement of the relative error between 2 curves. • f1= {[Σ t=1n |Rt-Tt|] / [Σ t=1n Rt]} ×100. • Values range from 0 to 15
  • 113. • Similarity Factor (F 2 value)-define as measurement of similarity in % Dissolution between two curve. • Where Rt and Tt = cumulative % dissolved for reference and test • Values range from 50 to 100 113
  • 114. USP SGF (simulated gastric fluid) NaCl 2.0 g Purified pepsin 3.2 g HCl 7.0 mL Purified water qs. 1000 mL Media has a pH of about 1.2 USP SIF (simulated intestinal fluid) Monobasic potassium phosphate 6.8 g in Purified water 250 mL NaOH (0.2 N) 77 mL and Purified water 500 mL Pancreatin 10.0 g Adjust with either 0.2 N NaOH or 0.2 N HCl to a pH of 6.8 ± 0.1. Purified water qs. 1000 mL
  • 115. Oral Drug Absorption Gastric Emptying Transit Permeation Dissolution Metabolism
  • 116. In Vivo and In Vitro Relationship: Scientific Issues • Limits to oral drug absorption – Dissolution-limited – Solubility-limited – Permeability-limited Gastric Emptying Transit Permeation Dissolution Metabolism SSoolluubbiilliittyy Dissolution Permeation Conc ³ Solubility
  • 117. IN VITRO IN VIVO CORRELATION
  • 118. In vitro-in vivo correlation • A predictive mathematical model that describes the relationship between an in-vitro property of a dosage form and an in-vivo response.
  • 119. • Key goal in development of dosage form and good understanding of in vitro and in vivo performance of dosage form • Formulation optimization requires altering some parameters – bioavailability studies • Regulatory guidance developed to minimize the additional bioavailability studies
  • 120. 120 Purpose of IVIVC • The optimization of formulations – may require changes in the composition, manufacturing process, equipment, and batch sizes. – In order to prove the validity of a new formulation, which is bioequivalent with a target formulation, a considerable amount of efforts is required to study bioequivalence (BE)/bioavailability(BA). • The main purpose of an IVIVC model – to utilize in vitro dissolution profiles as a surrogate for in vivo bioequivalence and to support biowaivers – Data analysis of IVIVC attracts attention from the pharmaceutical industry.
  • 121. Basic approaches • By establishing a relationship usually linear, between the in vitro dissolution and in vivo bioavailability parameters. • By using data from previous bioavailability studies to modify the dissolution methodology.
  • 122. • In vitro – in vivo correlation can be achieved using  Pharmacological correlation  Semi quantitative correlation  Quantitative correlation
  • 123. In vitro-in vivo correlations • Correlations based on the plasma level data • Correlations based on the urinary excretion data • Correlations based on the pharmacological response
  • 124. DEFINITION • USP definition “The establishment of rational relationship b/w a biological property or a parameter derived from a biological property produced by a dosage form and physicochemical property of same dosage form” • FDA definition “It is predictive mathematical model describing the relationship b/w in vitro property of dosage form and a relevant in vivo response”
  • 125. IMPORTANCE • Serves as a surrogate of in vivo and assist in supporting biowaivers • Validates the use of dissolution methods and specification • Assist in QC during mfg and selecting the appropriate formulation
  • 126. IVIVC levels • Level A: – Point to point correlation is developed between in vitro dissolution rate and the in vivo rate of absorption • Level B: – Utilises statistical moment analysis and the mean in vitro dissolution time is compared to either the mean residence time or the mean in vivo dissolution time • Level C: – single point correlation that relates one dissolution time point to one pharmacokinetic parameter Multiple level C
  • 127. Level A correlation • Highest category correlation • Represents point to point relationship • Developed by two stage procedure Deconvulation Comparison • Purpose – define direct relationship 120 100 80 60 40 20 0 0 20 40 60 80 100 120 % Drug Dissolved % Drug Absorbed
  • 128. Level B correlation • Utilizes the principle of statistical moment analysis MDT vitro is compared with MRT vivo • No point to point correlation • Does not reflect the actual in vivo plasma level curves
  • 129. Level C correlation • Dissolution time point (t 50%,t 90% ) is compared to one mean pharmacokinetic parameter ( Cmax ,tmax ,AUC) • Single point correlation • Weakest level of correlation as partial relationship b/w absorption and dissolution is established • Useful in the early stages of formulation development
  • 130. Multiple level C correlation • It reflects the relationship b/w one or several pharmacokinetic parameter of interest and amount of drug dissolved at several time point of dissolution profile • Base on  Early  Middle  Late stage
  • 131. Applications • Ensure batch to batch consistency • Serve as a tool in the development of a new dosage form with desired in-vivo performance • Assist in validating or setting dissolution specifications