You are on page 1of 208

BIOPHARMACEUTICS &

PHARMACOKINETICS
(BP 604T)
Unit 1
Introduction to Biopharmaceutics

MADHU VERMA
SYLLABUS
• Introduction to Biopharmaceutics
• Absorption: Mechanisms of drug absorption through
GIT, factors influencing drug absorption though GIT,
absorption of drug from Non per oral extra-vascular
routes,
• Distribution: Tissue permeability of drugs, binding of
drugs, apparent, volume of drug distribution, plasma
and tissue protein binding of drugs, factors affecting
protein-drug binding. Kinetics of protein binding,
Clinical significance of protein binding of drugs
Biopharmaceutics
• USP 1972 define the biopharmaceutics
It is the study of the factors influencing the
bioavailability of a drug in man and animals
and the use of this information to optimize
pharmacological and therapeutic activity of
drug products’’
Or The science that examines interrelationship
of the physicochemical properties of the drug,
the dosage form in which the drug is given,
and the route of administration on the rate
and extent of systemic drug absorption.
Introduction of Absorption
• Definition :
The process of movement of unchanged
drug from the site of administration to systemic
circulation.

 There always exist a correlation between the plasma


concentration of a drug & the therapeutic response
& thus, absorption can also be defined as the
process of movement of unchanged drug from the
site of administration to the site of measurement.
i.e., plasma.
Therapeutic success of a
rapidly & completely Not only the
absorbed drug. magnitude of drug
that comes into the
Plasma Minimum effective conc. systemic circulation
Drug
Therapeutic failure of a
but also the rate at
Conc. slowly absorbed drug. which it is absorbed
is important this is
Subtherapeutic level clear from the figure.

Time

5
CELL MEMBRANE €
 Also called the plasma membrane, plasmalemma or
phospholipid bilayer.
 The plasma membrane is a flexible yet sturdy barrier that
surrounds & contains the cytoplasm of a cell.

 Cell membrane mainly consists of:


1. Lipid bilayer-
-phospholipid
-Cholesterol
-Glycolipids.
2. Proitens-
-Integral membrane proteins
-Lipid anchored proteins
-Peripheral Proteins
LIPID BILAYER
• The basic structural framework of the plasma
membrane is the lipid bilayer.
• Consists primarily of a thin layer of amphiphilic
phospholipids which spontaneously arrange so that
the hydrophobic “tail” regions are shielded from the
surrounding polar fluid, causing the more hydrophilic
“head” regions to associate with the cytosolic &
extracellular faces of the resulting bilayer.
• This forms a continuous, spherical lipid bilayer app.
7nm thick.
It consists of two back to back layers made up of
three types: Phospholipid, Cholesterol, Glycolipids.

1) Phospholipids : Principal type of lipid in


membrane about 75 %.
Contains polar and non polar
region.
Polar region is hydrophilic and
non polar region is hydrophobic.
Non polar head contain two fatty
acid chain.
One chain is straight fatty acid
chain.( Saturated )
Another tail have cis double bond
and have kink in tail.
( Unsaturated )
CHOLESTEROL
• Amount in membrane is 20 %.
• Insert in membrane with same
orientation as phospholipids
molecules.
• Polar head of cholesterol is
aligned with polar head of
phospholipids.

FUNCTION:
• Immobilize first few
hydrocarbons groups
phospholipids molecules.
• Prevents crystallization of
hydrocarbons &
phase shift in membrane
OH
GLYCOLIPIDS
• Another component of membrane lipids present about 5 %.

• Carbohydrate groups form polar “head”.

• Fatty acids “tails” are non polar.

• Present in membrane layer that faces the extracellular fluid.

• This is one reason due to which bilayer is asymmetric.

• FUNCTIONS:
Protective
Insulator
Site of receptor binding
COMPOSITION OF PROTEINS

PROTEINS

LIPID
INTEGRAL PERIPHERAL
ANCHORED
PROTEINS PROTEINS
PROTEINS
INTEGRAL PROTEINS
• Also known as “Transmembrane protein”.
• Have hydrophilic and hydrophobic domain.
• Hydrophobic domain anchore within the cell
membrane and hydrophilic domain interacts with
external molecules.
• Act as carrier for transporting the substances
• Ex. – Ion Channels, Proton pump, GPCR.
LIPID ANCHORED PROTEIN

• Covalently bound to single or multiple lipid


molecules.

• The protein itself is not in contact with membrane.

• Ex. – G Proteins.
PERIPHERAL PROTEINS
• Attached to integral membrane proteins OR associated
with peripheral regions of lipid bilayer not penetrate .

• Have only temporary interaction with biological


membrane.

• Once reacted with molecule, dissociates to carry on its


work in cytoplasm.

• Ex. – Some Enzyme, Some Hormone


MECHANISM OF DRUG ABSORPTION
A) Transcellular /intracellular transport
Passage of drugs across GI epithelium, most common
pathway.
1) Passive transport: no need of energy
eg. Passive diffusion , Pore transport, Ion-pair transport,
mediated or Facilitated diffusion

2) Carrier- mediated Active transport : need of energy


eg. A. Primary Active transport
B. Sec. Active transport ie symport and antiport
B) paracellular / intercellular transport:
Passage of drugs through the junctions b/t GI
epithelium cells or persorption

C) vesicular transport or corpuscular transport


(endocytosis): energy dependent,
transport across the cell membrane (with in the
vesicles)
Eg. Pinocytosis, phagocytosis
1) PASSIVE DIFFUSION

• Also known as non-ionic


diffusion.
• Absorption of 90% of drugs.
• The driving force for this
process is the concentration
or electrochemical gradient.
It is defined as the
difference in the drug
concentration on either
side of the membrane.
• Passive diffusion is best expressed by Fick’s first
law of diffusion which states that the drug
molecules diffuse from a region of higher
concentration to one of lower concentration until
equilibrium is attained & the rate of diffusion is
directly proportional to the concentration
gradient across the membrane.

dQ = D A Km/w (CGIT – C)
dt h

Characteristic of passive diffusion:


a) Down hill transport, energy independent
b) Greater the surface area & lesser the thickness of the
membrane, faster the diffusion.
c) Equilibrium is attained when the concentration on either
side of the membrane become equal.
d) Greater the membrane/ water partition coefficient of
drug, faster the absorption.
e) Unionized drug absorbed
f) Drug diffuse rapidly in case of low GI fluid. On dilution,
diffusion is reduced due to reduction in concentration
gradient.
 Passive diffusion process is energy independent but
depends more or less on the square root of the
molecular size of the drugs.
 The mol. Wt. of the most drugs lie between 100 to 400
Daltons which can be effectively absorbed passively.
• Initially Cgit ˃˃˃ C, larger con gradient--------faster abs
• DQ/dt = PCgit
• Where P = permeability coefficient

WHY ????????
• Why absorption rate is always more than elimination?
• Why the concentration of drug at the site of absorption
(Cgit) is usually much greater than on the other side of the
membrane ?
• This is because of the rapid dilution of the drug in the blood
and its subsequent distribution to the tissues.
2) Pore transport
• Also known as convective transport, bulk flow or
filtration.
• Important in the absorption of low mol. Wt. (less than
100). Low molecular size (smaller than the diameter of
the pore) & generally water-soluble drugs through
narrow, aqueous filled channels or pores in the
membrane structure. Chain or linear drug up to 400 D
can absorbed easily
e.g. urea, water & sugars.

 The driving force for the passage of the drugs is the


hydrostatic or the osmotic pressure difference across
the membrane.
• The rate of absorption via pore transport depends on
the number & size of the pores, & given as follows:

dc = N. R2. A . ∆C
dt (η) (h)
where,
dc = rate of the absorption.
dt
N = number of pores
R = radius of pores
∆C = concentration gradient
η = viscosity of fluid in the pores
3) CARRIER MEDIATED TRANSPORT
MECHANISM
• Involves a carrier (a component of the membrane)
which binds reversibly with the solute molecules to
be transported to yield the carrier solute complex
which transverses across the membrane to the other
side where it dissociates to yield the solute molecule
• The carrier then returns to its original site to accept a
fresh molecule of solute.
• Carriers may be transporter protein or enzymes
Characteristics of carrier mediated transport

• A carrier is uncharged or nonpolar molecule


• No direction-------work in both direction
• Structure specific (false nutrients)
• Saturable process
• Nonlinear pharmacokinetics
• There are two types of carrier mediated transport
system:
a)facilitated diffusion
b) active transport
a) Facilitated diffusion

• This mechanism involves


the driving force is
concentration gradient.
• In this system, no
expenditure of energy is
involved (down-hill
transport), therefore the
process is not inhibited by
metabolic poisons that
interfere with energy
production.
• Limited importance in the absorption of drugs.
e.g. entry of glucose into RBCs & intestinal absorption
of vitamins B1 & B2.
 A classical example of passive facilitated diffusion is
the gastro-intestinal absorption of vitamin B12.
 An intrinsic factor (IF), a glycoprotein produced by the
gastric parietal cells, forms a complex with vitamin B12
which is then transported across the intestinal
membrane by a carrier system.
b) Active transport
• Requires energy from ATP
• It may be
• A) primary Active transport: direct need of ATP. Eg.
absorption of glucose. again two type
i) Ion transporters: eg. Na+/K+ ATPase, H+/ k+ ATPase, Ca++
ATPase
ii) ATP binding cassette (ABC) transporters: transporting out
the cell, exsorption. P-glycoprotein act this work. Responsible
for MDR
B)Secondary active transport : no direct requirement of
ATP. It may be-
Symport: Na+ glucose symporters, H+ copuled peptide transporter eg.
Beta- lactam antibiotic
Antiport : (counter transport): expulsion of H+ using Na+ gradient in
kidney
Active transport cont…………
Characteristics of Active transport
• More important process than
facilitated diffusion.
• The driving force is against
the concentration gradient
or uphill transport.
• Since the process is uphill,
energy is required in the
work done by the barrier.
• As the process requires
expenditure of energy, it can
be inhibited by metabolic
poisons like fluoride, cyanide
and dinitrophenol etc. that
interfere with energy
production.
• If drugs (especially used in cancer) have structural similarities
to such agents, they are absorbed actively.
• A good example of competitive inhibition of drug absorption
via active transport is the impaired absorption of levodopa
when ingested with meals rich in proteins.

• Mixed order kinetics: The rate of absorption by active


transport can be determined by applying the equation used
for Michalies-menten kinetics:
dc = [C].(dc/dt)max
dt Km + [C]
Where,
(dc/dt)max = maximal rate of drug absorption at high drug
concentration.
[C] = concentration of drug available for absorption
Km = affinity constant of drug for the barrier.
5) ION PAIR TRANSPORT

• It is another
mechanism is
able to explain
the absorption
of such drugs
which ionize at
all pH condition.
like qu. Amm
comp,
tetracyclines
• Transport of charged molecules due to the formation
of a neutral complex with another charged molecule
carrying an opposite charge.
• Drugs have low o/w partition coefficient values, yet
these penetrate the membrane by forming reversible
neutral complexes with endogenous ions.
e.g. mucin of GIT.
• Such neutral complexes have both the required
lipophilicity as well as aqueous solubility for passive
diffusion and cross lipoidal membrane.
• This phenomenon is known as ion-pair transport.
• Eg. Propranolol attached with oleic acid
6) ENDOCYTOSIS
• It involves engulfing
extracellular materials within a
segment of the cell membrane
to form a saccule or a vesicle
(hence also called as
corpuscular or vesicular
transport) which is then
pinched off intracellularly.
• No need of aq. Solubility of
drug.
• Responsible for fat and waters
soluble vit like B12, insulin
• Absorb by lymphatic
circulation ---avoid fist pass
effect
• In endocytosis, there are three process:

A) Phagocytosis

B) Pinocytosis

C) Transcytosis
A) Phagocytosis
B) Pinocytosis

• This process is
important in the
absorption of oil
soluble vitamins & in
the uptake of
nutrients, protein,
sabin polio vaccine.
C) Transcytosis

• It is a phenomenon in which endocytic vesicle


is transferred from one extracellular
compartment to another.
Phases of drug transfer from GI to
systemic circulation
&
First pass metabolism
Phases of drug transfer from GI to systemic circulation

Three phases
1. Pre systemic phase
a. dissolution of drug
b. metabolism of drug by digestive enzyme &
bacterial enzyme in colon
2. UP Take Phase: three process involved
a. Delivery of drug at absorption site
b. Metabolism by GI epithelium (gut wall
enzyme)
c. Passage of drug through GI epithelium
28-Mar-20 42
3. Post uptake phase: three phases
a. Metabolism liver (first pass hepatic
metabolism)
b. Enterohepatic circulation of drug: drug may
excreted in bile, re-enter to GIT via gall
bladder & gets reabsorbed
c. Transfer of drug into systemic circulation
Route of drug transfer from absorption site to
in to systemic circulation:
a. Splanchnic circulation: major route of absorption
1. The drug then is absorbed in the GIT and enters
the portal circulation before entering the systemic
circulation. Via the portal circulation it enters the
liver where some drugs undergo
extensive biotransformation and the drug
concentration is decreased.
2. This happens most commonly through oral intake
eg. Propanolol, Lidocaine, Diazepam

28-Mar-20 44
2. Lymphatic circulation
• Minor pathway of absorption : due to
a. rate of lymph flow in GIT is approximately 500 times less than
that of blood flow in the portal vein
b. Lymph vessels are less accessible than capillaries
Eg . Fat, fat soluble vitamins, lipophilic drugs and drugs having
high molecular wt (above 16000 d)

Advantages :
a. avoidance of first pass metabolism
b. direct delivery of drugs to particular regions of the lymphatic
circulation e.g. in the treatment of disease states
First-pass effect or Pre systemic metabolism/ First pass
metabolism
when an administered drug enters the liver and undergoes
extensive biotransformation and thus decreasing the
concentration rapidly before it reaches in to systemic circulation or
its target.

Four primary systems which affect pre systemic


metabolism of a drugs.
Luminal enzymes I e digestive enzymes
Gut wall enzymes or mucosal enzymes.
Bacterial enzymes.
Hepatic enzymes.

28-Mar-20 46
• Luminal enzymes (digestive enzyme): These enzymes present
in gut fluids and include enzymes from intestinal and
pancreatic secretions.
Eg. Hydrolases… hydrolyse ester drugs like chloramphenical
palmitate into chloramphenical (active)
peptidases split amide linkage of polypeptide/ protein drugs
….inactive them

• Gut wall enzymes: Also called mucosal enzymes they are


present in gut and intestine, colon mucosa.
• Eg ADH present stomach mucosa inactivates ethanol.
Intestinal and colonic mucosa contain both phase I and Phase
II enzyme
• However the enzymes of the proximal small intestine are
most active

28-Mar-20 47
• Bacterial enzymes: GI microflora scantily present in
stomach and small intestine and is rich in colon.
Most of drug are unaffected.
Sulfasalazine------Sulfamyridine + 5 ASA by colonic
enzyme

• Hepatic enzyme: several drug undergo first-pass


hepatic metabolism,
• isoprenaline, nitroglycerin, morphine, diazepam,
lidocaine etc.
FACTOR AFFECTING DRUG
ABSORPTION

3/28/2020 50
Factor affecting drug absorption
Biopharmaceutical consideration in dosage form
• For absorption rate and extent both important
• Rate at which the drug reaches the systemic circulation is
determined by slowest step among the various steps involved
in sequence
• Sequence of events that take place like
1. Disintegration
2. Disaggregation
3. Dissolution
4. Permeation (absorption)

3/28/2020 51
Following factors affects the abs.

A. pharmaceutical factors
1. physiochemical properties
i. drug solubility and dissolution
ii. Particles size and surface area
iii. Polymorphism and amorphism
iii. Pseudo polymorphism ( hydrates/ solvates)
iv. Salt form of drug
v. lipid solubility, Pka, pH- partition hypothesis
iv. Drug stability
2. Dosage form related factors including excipients
B. Patient related factors
Age, gastric emptying time, intestinal transit time, gastrointestinal pH, disease
state, blood flow through GIT, gastrointestinal content, presystemic
3/28/2020 52
metabolism
2. Dosage form related factors including
excipients
i. Disintegration time
ii. Dissolution time
iii. Manufacturing variables
iv. Nature and type of dosage
v. Storage condition
B. Patient related factors
Drug solubility and dissolution rate
Definition-
Solubility: max amt of drug dissolved in given solvent under
the std condition of temp, pressure & pH. Static property
Drug with solubility less than 1 mg/ml………problematic
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.

3/28/2020 54
BCS classification: Amidon et al classified the drugs
on the bases of solubility & permeability

• Class 1: High Solubility-High Permeability: generally very well-


absorbed compounds eg ditiazem
• Class 2: Low Solubility-High Permeability: exhibit dissolution
rate-limited absorption. Eg nifidipine
• Class 3: High Solubility-Low Permeability: exhibit permeability
rate-limited absorption. Eg insulin
• Class 4: Low Solubility-Low Permeability: very poor oral
bioavailability. Eg. taxol

3/28/2020 55
• HIGHLY SOLUBLE when the highest dose strength is soluble
in≤250 ml water over a pH range of 1 to 7.5.
• HIGHLY PERMEABLE when the extent of absorption in
humans is determined to be ≥90% of an administered dose,
in comparison to an intravenous reference dose.
• RAPIDLY DISSOLVING when ≥85% of labelled amount of drug
substance dissolves within 30 minutes using USP apparatus I
or II in a volume of ≤900 ml buffer solutions

• The pH-solubility profile of the test drug substance should be


determined in aqueous media with a pH in the range of 1-7.5 using
traditional shake-flask method as well as acid or base titration methods .

3/28/2020 56
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.

3/28/2020 57
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.
3/28/2020 58
3/28/2020 59
• The rate of dissolution is given by Noyes and
Whitney:
dc
= k (Cs- Cb)
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
3/28/2020 60
Modified Noyes-Whitney’s Equation -

dC = DAKw/o (Cs – Cb )
dt 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.
3/28/2020 61
• 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.
• In vitro sink condition can be maintain if Cb is always
less than 10 % of Cs

• 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.

3/28/2020 62
• Under sink conditions, if the volume and surface area
of the solid are kept constant, then

dC
= K
dt

• This represents that the dissolution rate is constant


under sink conditions and follows zero order kinetics.

3/28/2020 63
Dissolution rate under non-sink and
sink conditions.

zero order dissolution


Conc. of dissolved drug under sink condition
first order dissolution
under non-sink
condition

Time

3/28/2020 64
• Hixon-Crowell’s cubic root law of dissolution
takes into account the particle size decrease
and change in surface area,

W01/3 – W1/3 = Kt
Where,
W0=original mass of the drug
W=mass of drug remaining to dissolve at time t
Kt=dissolution rate constant.
3/28/2020 65
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.

3/28/2020 66
3/28/2020 67
• The Danckwert’s model is expressed by equation

dC = dm = A (Cs-Cb). γD
V dt
dt
Where,
m = mass of solid dissolved
Gamma (γ) = rate of surface renewal

3/28/2020 68
III. Interfacial barrier model/Double barrier or Limited
solvation theory :-
based on two assumption:
1. RDS in dissolution is mass transport.
2. solid solution equilibrium is achieved at solid liquid interface
an intermediate con. can exist at interface as a result of
solvation mechanism and is a function of solubility rather
than diffusion.
• 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.
3/28/2020 69
Factors affecting Drug Dissolution or bioavailability of
drug

A. Factors relating to the physicochemical properties


of drug.

B. Factors relating to the dosage forms.

3/28/2020 70
A. Factors relating to the physicochemical
properties of drug-
i. Solubility-
• Solubility plays important role in controlling dissolution
from dosage form.
• From Noyes-Whitney equation it shows that aqueous
solubility of drug which determines its dissolution rate.

• Dissolution rate (R) can be calculated from its saturation


solubility by formula
R =Dc/dt = 2.24Cs

To avoid Bio availability problem minimum aqs solubility should


be 1 %.
3/28/2020 71
The Solution Process Model
• Whether or not a substance will dissolve in another
substance depends on the magnitude of the forces
between molecules.
 Cohesive forces: Forces between similar molecules
 Adhesive forces: Forces between dissimilar molecules

Solute molecules Solvent molecules Solute + solvent


cohesive cohesive adhesive

3/28/2020 72
The Solution Process Model
• Step # 1:
• Removal of solute molecule from the pure solute
structure, this results in loss of energy due to breakage of
cohesive forces.

3/28/2020 73
The Solution Process Model
• Step # 2:
• Creation of a hole in solvent molecules by disrupting
intermolecular forces between solvent molecules. This
results in loss of energy due to breakage of cohesive
forces:

3/28/2020 74
The Solution Process Model
• Step # 3:
• Introduction of the solute molecule into the hole in the
solvent structure and subsequent solvation of the solute
molecule by the solvent. This results in gain of energy
from formation of new adhesive forces :

In general:
Overall gain of energy < loss of energy, solubility will be very
high.
Overall gain of energy > loss of energy, solubility will be very
3/28/2020
low. 75
ii. Particle size and effective surface area of the
drug –
• Particle size and surface area are inversely related to each
other. Surface area directly related to dissolution rate.
• Smaller particle size, greater surface area then higher will
be dissolution rate, because dissolution is thought to take
place at the surface area of the solute
( Drug).
Two types of surface area –
Absolute surface area which is the total surface area of any
particle.
Effective surface area which is the area of solid surface
exposed to the dissolution medium.
Particle size may be reduce by micronization

3/28/2020 76
• Effective surface area is directly related to the dissolution
rate.

• Greater the effective surface area, more intimate the contact


between the solid surface and the aqueous solvent and faster
the dissolution but it is only when micronization reduce the
particle size below 0.1 micron resulting increase intrinsic
solubility------increase dissolution rate
• This is true only in case of hydrophilic drug. Eg
Griesiofulvin (half dose) Spironolactone (20 times dose),
Digoxin (100 % BA).

This is due to higher energy of small particles than bulk of solid


resulting increase interaction with solvent.

3/28/2020 77
In case HYDROPHOBIC DRUGS micronisation techniques
results in decreased effective surface area & thus fall in
dissolution rate.
eg. Micronization of Aspirin, phenobarbital, lesser effective
surface area and hence lesser dissolution rate

REASON FOR THESE:


1) The hydrophobic surface of the drugs adsorbs air on to their
surface which inhibits their wettability.
2) The particles reaggregate to form large particles due to their
high surface free energy , which either float on the surface or
settle on the bottom of the dissolution medium.
3) Electrically induced agglomeration owing to surface charges
prevents intimate contact of the drug with the dissolution
medium.
3/28/2020 78
For such hydrophobic drugs absolute SA can be converted to
their effective surface area by addition of Surfactant or
hydrophilic polymer like PVP,PEG, dextrose .
Molecular and solid dispersion are other technique to improve
the solubility.

• Finally drug size reduction and subsequent increase in surface


area and dissolution rate is always not useful.
• Ex. Penicillin G & Erythromycin
• These Drugs are unstable and degrade quickly in solution.
• Sometime, reduction in particle size of nitrofurantoin and
piroxicam increase gastric irritation
• These problem can be overcome by Microencapsulation.

3/28/2020 79
iii. Polymorphism and amorphism –
• When a substance exists in more than one crystalline form,
the different forms are designated as polymorphs and the
phenomenon as Polymorphism.
Eg. chloramphenicol palmitate, cortisone acetate, tetracyclines,
sulphathiazole and paracetamol.
It May be
• Enantiotropic: reversibly changes from one form to another by
temp & pressure eg sulphur
• Monotropic: unstable at all temp & pressure eg. Glyceryl
monostearate
• Polymorph differ each other in their physical characters like
solubility, MP, density, hardness, compression characteristics.
• Identified by OC, XRD, DSC.

3/28/2020 80
• Depending upon stability polymorph may be
• Stable polymorphs has lower energy state, higher M.P. and
least aqueous solubility.
• Metastable polymorphs has higher energy state, lower M.P.
and higher aqueous solubility.
• Metastable form show grater bioavalability than stable form
(eg. Chloamphanical A, B, C but B is best in BA, riboflavin II
show 20 times more solubility than I, )
• Metastable form are thermodynamically unstable. Upon
storage metastable may converted to stable eg. Cortisone
acetate II convert to less soluble form V resulting caking of
solid.

• This change can be preventing by dehydrating the molecular


environment or by adding viscosity building agent like PVP,
CMC, pectin, gelatin
3/28/2020 81
• 40 % drug are polymorphic but barburates show highest 70%
and sulphonamides 65 % in polymorphic form
• Amorphous form of drug which has no internal crystal
structure represents higher energy state and greater
aqueous solubility than crystalline forms.
• E.g.- amorphous form of novobiocin is 10 times more
soluble than the crystalline form. Other eg. includes
chloramphenical palmitate, cortisone acetate,
phenobarbital exhibit higher water solubility.
• Thus, the order for dissolution of different solid forms
of drug is –
amorphous > metastable > stable

3/28/2020 82
iv. Hydrates/solvates –
• The stoichiometric type of adducts where the solvent
molecules are incorporated in the crystal lattice of the solid are
called as the solvates & trapped solvent is known as solvent of
crystallization.
• Solvates can exist in different crystalline from is known as
pseudopolymorphism.
• When the solvent in association with the drug is water, the
solvate is known as hydrate.
• Hydrates are most common form of solvate
• The organic solvates have greater aqueous solubility than the
nonsolvates.
• E.g. – chloroform solvates of griseofulvin is more water soluble
than their nonsolvated forms

3/28/2020 83
• Generally anhydrous form r more water
soluble than hydrates because hydrates r
already in interaction with water & therefore
have less energy for crystal break up.
• Eg. Anhy. theophylline, ampicillin r more
soluble than monohydrate and trihydrate
respctively.

• Solvates are differ only their physical


properties.

3/28/2020 84
IV. Salt form of the drug-

• Dissolution rate of weak acids and weak bases can


be enhance by converting them into their salt form.
• ex. Na salts of Novobiocin shows improved bioavailability
• With weakly acidic drugs, a strong base salt is
prepared like sodium and potassium salts of
barbiturates and sulfonamides.

• With weakly basic drugs, a strong acid salt is prepared like


the hydrochloride or sulfate salts of alkaloidal drugs.

85
• Influence of salt formation on drug solubility , dissolution rate,
& abs. can be explain by considering pH of diffusion layer not
the pH of bulk solution.

• Eg salt of weak acid pH of diffusion layer is slightly more than


bulk sol resulting increase in solubility.
• Salt of weak base pH of diffusion layer is slightly less than bulk
sol resulting increase in solubility.
• This is due to the buffering action of strong base cation and
strong acid anion respectively.

• Principal of in situ salt formation is utilized to enhance


solubility of drug like aspirin, penicillin. This also reduced GI
irritation & poor hydrolytic stability of aspirin

86
• size of counter ions:
• Eg. Novobiocin sod & calcium, free acid(50:
25:1)
• Generally counter ions are larger size have
poor ionic strength, poor solubility.
• Eg. Pamoates, stearates, palmitates of weak
bases have poor aqu. Solubility.
• These complex are useful to prolongs duration
of action, taste masking, enhance the GI
stability

87
Drug pKa, lipophilicity and GI pH i.e. pH
PARTITION THEORY (Brodie theory) :

It explain drug absorption from GIT and its distribution


across biomembranes.

Drug(>100 daltons) transported by passive diffusion


depend upon:

 dissociation constant, pKa of the drug


 lipid solubility, K o/w
 pH at absorption site.

Most drugs are either weak acids or weak bases whose


degree of ionization is depend upon pH of biological fluid.
For a drug to be absorbed, it should be unionized and the
unionized portion should be lipid soluble.

The fraction of drug remaining unionized is a function of both


Dissociation constant (pKa) and pH of solution.

• The above statement of hypothesis is based on the


assumptions that;
• The GIT is a simple lipoidal barrier for drug transport
• Larger fraction of unionized drug faster absorption.
• Grater lipophilicity (Ko/w) of unionized drug better abs.

89
pKa of the drug and pH of GIT:
Henderson Hasselbatch Equation
For acid,
pH = pKa + log[ ionised drug/unionized drug ]

% drug ionised = 10 (pH – pka) /1+ 10 (pH – pka)

For base,
pH = pKa + log[un ionized drug/ionized drug ]

% drug ionised = 10 (pka –pH) /1+ 10 ((pka –pH)

Eg. Weak acid aspirin (pKa=3.5) in stomach (pH=1) will have > 99%of
unionized form so gets absorbed in stomach

Weak base quinine (pKa=8.5) will have very negligible unionization in


gastric pH so negligible absorption

10
Several prodrugs have been developed which are lipid
soluble to overcome poor oral absorption of their parent
compounds.

eg. Pivampicilin, the pivaloyloxy-methyl ester of ampicilin


is More lipid soluble than ampicilin.

91
INFLUENCE OF DRUG pKa AND GI pH ON DRUG
ABSORBTION
Drugs Site of absorption
Very weak acids (pKa > 8.0) Unionized at all pH values
Absorbed along entire length of GIT
Moderately weak acids (pKa 2.5 – 7.5) Unionized in gastric pH
Ionized in intestinal pH
Better absorbed from stomach
Strong acids (pKa <2.5) Ionized at all pH values
Poorly absorbed from git
Very weak bases (pKa < 5) Unionized at all pH values
Absorbed along entire length of GIT
Moderately weak bases (pKa 5 – 11 ) Ionized in gastric ph
Unionized in intestinal pH
Better absorbed from intestine
Strong bases (pKa >11) Ionized at all ph values
Poorly Absorbed from GIT

92
PHARMACEUTICAL FACTORS
1) Disintegration time (tablets/capsules):
Rapid disintegration is important to have a rapid absorption so
lower disintegration time is required.

Disintegration time of tablet is directly proportional to –amount of


binder & Compression force.

E.g. COATED TABLETS: they have long disintegration time.


Fast dispersible tablets have short disintegration time

In vitro disintegration test gives no means of a guarantee of drugs


bioavailability because if the disintegrated drug particles do not
dissolve then absorption is not possible.
2) Dissolution time:
Dissolution is a process in which a solid substance solubilises in a
given solvent i. e… mass transfer from the solid surface to the
liquid phase.

Dissolution time is also an important factor which affect the drug


absorption.

3) Manufacturing variables:
Several manufacturing processes influence drug dissolution from
solid dosage forms.

For example: For tablet it is


Presence of excipient
Method of granulation
Compression force
Method of granulation:
The wet granulation process is the most conventional technique
The tablets that dissolve faster than those made by other
granulation methods.
But wet granulation has several limitations like formation of
chemical degradation like- hydrolysis.
The method of direct compression force has been utilized to yield
the tablets that dissolve at a faster rate.
Compression force:
The compression force employed in tabletting process influence
density, porosity, hardness, disintegration time and dissolution rate
of tablets.
Higher compression force increases the density and hardness of
the tablet, decreases porosity and hence penetrability of the solvent
into the tablet and thus in slowing of dissolution and absorption
(Fig .A)
On the other hand, higher compression force causes deformation,
crushing or fracture of drug particles into smaller ones and causes a large
increase in effective surface area. This results in an increase in
dissolution rate of tablets (Fig B)

A combination of both the curves A and B is also possible as shown in


curves C & D.

Fig.Influence of compression force on the dissolution rate of tablets


4) Pharmaceutical ingredients (excipients/adjuvants):
More the number of Excipients in the dosage form, more complex
it is & greater the potential for absorption and Bioavailability
problems.
Commonly used excipients in various dosage forms are,

a) Vehicle:
Rate of absorption – depends on its miscibility with biological
fluid.
Aqueous vehicle: water, syrup
Non aqueous miscible vehicles causes rapid absorption e.g.
propylene glycol.
Immiscible vehicles – Absorption depends on its partitioning from
oil phase to aqueous body fluid.
b) Diluents
Hydrophilic diluents – Imparts Absorption e.g starch, lactose
Hydrophobic diluents – Retards Absorption e.g
Also, there is a drug-diluent interaction, forming insoluble
complex and retards the absorption. E.g. Tetracycline-DCP

c) Binders & granulating agent: hold powder together


Hydrophilic binders – Imparts hydrophilic properties to the
granule surface – gives better dissolution properties. E.g. Starch,
Gelatin. PVP.
More amount of binder increases the hardness of the tablet and
retards the absorption rate.
Non aqueous binders: EC- retard drug dissolution

d) Disintegrants
Mostly hydrophilic in nature.
Decrease in amount of disintegrants – significantly lowers
bioavailability.
e) Lubricants:
Commonly hydrophobic in nature – therefore inhibits penetration of water into
tablet and thus dissolution and disintegration. Eg stearates,
SLS and carbowaxes soluble lubricants….increase dissolution

f) Suspending agents/viscosity agent:


Stabilized the solid drug particles and thus affect drug absorption.
Macromolecular gum forms un-absorbable complex with drug e.g. Na CMC.

Viscosity imparters – act as a mechanical barrier to diffusion of drug from its


dosage form and retard GI transit of drug.

g) Surfactants:
May enhance drug absorption by interacting with drug or membrane or both.
e.g. Griseofulvin, steroids
It may decrease absorption when it forms the un-absorbable complex with drug
above CMC.
h) Coating:
In general, deleterious effects of various coatings on the drug
dissolution from a tablet dosage form are in the following order.
Enteric coat > sugar coat > non-enteric coat.

The dissolution profile of certain coating materials change on


aging; e.g. shellac coated tablets, on prolonged storage, dissolve
more slowly in the intestine. This can be however, be prevented by
incorporating little PVP in the coating formulation.

i) Buffers:
Buffers are sometimes useful in creating the right atmosphere for
drug dissolution as was observed for buffered aspirin tablets.
However, certain buffer systems containing potassium cations
inhibit the drug absorption as seen with Vitamin B2 and
sulfanilamide.
j) Colorants:
Even a low concentration of water soluble dye can have an
inhibitory effect on dissolution rate.
The dye molecules get absorbed onto the crystal faces and inhibit
the drug dissolution.
For example: Brilliant blue retards dissolution of sulfathiazole.

k) Complexing agents:
Complex formation has been used to alter the physicochemical &
biopharmaceutical properties of a drug.
Example
1)Enhanced dissolution through formation of a soluble complex.
E.g. ergotamine tartarate-caffeine complex & hydroquinone-
digoxin complex.
2)Enhanced lipophilicity for better membrane permeability.
E.g. caffeine-PABA complex.
5) Nature & type of dosage form:
Apart from the proper selection of the drug, clinical success often
depends to a great extent on the proper selection of the dosage form
of that drug.
As a general rule, the bio-availability of a drug form various
dosage forms decrease in the following order:
Solutions > Emulsions > Suspensions > Capsules > Tablets >
Coated Tablets > Enteric Coated Tablets > Sustained Release
Products.
6) Product age & storage condition:
Product aging and storage conditions can adversely affect the
bio-availability by change in especially the physico-chemical
properties of the dosage forms.

For example:
1.Precipitation of the drug in solution
2.Hardening of tablet
3.Change in particle size of suspension.
SOLUTIONS

Shows maximum bioavailability and factors affecting


Absorption from solution are as follows

1. Chemical stability of drug


2. Complexation: between drug and exipients of formulation
to increase the solubility, stability.
3. Solubilization: incorporation of drug into micelles to
increase the solubility of drugs.
4. Viscosity
5. Type of solution: Whether aqueous or oily solution.
SUSPENSIONS:
It comes next after solutions with respect to bioavailability
Factors that affects absorption from suspensions are

1. Particle size and effective surface area of dispersed phase

2. Crystal form of drug: some drug can change their crystal


structure.
Eg. Sulfathiazole can change its polymorphic form, it can be
overcome by addition by adding PVP.

3. Complexation: Formation of non absorbable complex between


drug and other ingredients.
Eg. Promazine forms a complex with attapulgite.
4. Inclusion of surfactant
Eg. The absorption of phenacitin from suspension is increased in
presence of tween 80.

5. Viscosity of suspension
Eg. Methyl cellulose reduces the rate and absorption of
nitrofurantoin

6. Inclusion of colourants:
Eg. Brilliant blue in phenobarbitone suspension.
CAPSULES

Two types of capsule

1. Hard gelatin capsule

2. Soft gelatin capsule


HARD GELATIN CAPSULE
The rate of absorption of drugs from capsule is function
Of some factors.
1.Dissolution rate of gelatin shell.
2.The rate of penetration of GI fluids into encapsulated mass
3.The rate at which the mass disaggregates in the GI fluid
4. The rate of dissolution.
5. Effect of excipients;
a).Diluents
b).Lubricants
c). Wetting characteristics of drug
d).Packing density
SOFT GELATIN CAPSULE
SGS has a gelatin shell thicker than HGS,but shell is
Plasticized by adding glycerin,sorbitol.SGS may used
To contain non aqueous solution or liquid or semi solid.

SGC have a better bioavailability than powder filled HGC


And are equivalent to emulsions.

Eg. Quinine derivative was better absorbed from SGC


Containing drug base compared with HGC containing
HCl salts.

Grieseoflavin exhibited 88% absorption from soft gelatin


Capsules compared to HGC(30%)
TABLETS

1.Compressed tablets

2. Coated tablets
Compressed tablets
Bioavailability are more due to large reduction
in surface area.

A B
Intact tablets a granules primary drug particles
K2
K1 K3
Drug in GI fluid

K4

Drug absorbed in body


The rate constants decrease in the following order.

K3>>K2>>K1

The overall dissolution rate and bioavailability of a poor


Soluble drugs is influenced by
1.The physicochemical properties of liberated particles.
2. The nature and quantity of additives.
3. The compaction pressure and speed of compression.
4. The storage and age of tablet
COATED TABLETS:
There are three types of coating
Sugar coating
Film coating
Enteric coating

SUGAR COATING:
Sugar,Shellac,fatty glycerides, bees wax, silicone resin
Sub coating agent: Talc,acacia,starch.

FILM COATING:
Polymers, dispersible cellulose derivatives like HPMC
CMC.

ENTERIC COATING:
Shellac, cellulose acetate phthalate etc.
Physiological factors or patient related
factors
1.Age
2.Gastric Empting
3.Intestinal Transit
4.GI pH
5.Blood Flow to GIT
6.Diseased State
7.GI Content
8.First pass effect

28-Mar-20 115
Gastrointestinal (GI) Physiology
pH Membrane Blood Supply Surface Area Transit Time By-pass liver
BUCCAL approx 6 thin Good, fast small Short unless yes
absorption with controlled
low dose

ESOPHAGUS 6 Very thick, no - small short -


absorption

STOMACH 1–3 Normal 0.15 L/min small 60- 120 minutes, no


Lipophilic,acidic reduced absorption
and neutral drugs

DUODENUM 5–7 Normal good large very short (6" no


Mainly lipohilic long)
and neutral drugs

SMALL 6 -7 Normal 1 L/min very large 10 - 14 about 3 hours no


INTESTINE All types of drugs ft, 200 cm2

LARGE 6.8 - 7 - good not very large 4 - long, up to 24 hr lower colon,


INTESTINE 5 ft rectum yes
Stomach is not principal region for absorption:

• Total mucosal surface area is small


• Epithelium is dominated by mucosal secreting cells rather
than absorptive cells
• Gastric residence time is limited
SMALL INTESTINE :
• Major site for absorption of most drugs due to its large surface area
(0.33 m2 ).
• It is 3-5 meters in length and is approximately 2.5-3 cm in diameter.
• The Folds in small intestine called as folds of kerckring, result in 3 fold
increase in surface area ( 1 m2).
• These folds possess finger like projections called Villi which increase
the surface area 30 times ( 10 m2).
• From the surface of villi protrude several microvilli which increase the
surface area 600 times ( 200 m2).
• Blood flow is 6-10 times that of stomach.
• PH Range is 5–7.5 , favourable for most drugs to remain unionised.
• Peristaltic movement is slow, while transit time is long.
• Permeability is high.

All these factors make intestine the best site for absorption of most drugs.
1. Age
In infants, the gastric pH is high and intestinal surface and blood flow to the
GIT is low resulting in altered absorption pattern in compare to adults.

In elderly persons, gastric emptying altered, decreased intestinal surface area
and GI blood flow, higher incidents of achlorhydria so impaired drug
absorption.

2. Gastric emptying time:


The process by which food leaves the stomach and enters the duodenum.
It is a RDS in drug absorption specifically intestinal absorption
Rapid gastric emptying is advisable where:
• Rapid onset action is required, eg; sedatives.
• Dissolution of drug occurs in intestine.
eg; Enteric coated tablets.
• Drug is unstable in gastric fluids. Penicillin G, Erythromycin
• Drug is best absorbed from distal part of small intestine,
• eg vitamin B 12 .
Delayed gastric emptying is required when drugs are absorbed
from proximal part of the small intestine and prolonged drug
absorption site contact is desired.

Gastric emptying is a first order process.

Gastric emptying rate: This is the speed at which the


stomach contents empty into the intestine.

Gastric emptying time: Which is the time required for the


gastric contents to the SMALL INTESTINE.

Gastric emptying half-life: which is the time taken for half


the stomach contents to empty.
Factors affecting GI emptying

1.Volume of meals
2.Composition of meal
3.Physical state of meal
4.GI pH
5.Body posture
6.Emotional state
7.Exercise
8.Drugs

28-Mar-20 121
1.Volume of meal: Larger the bulk of the meal, longer
the gastric time and however an initial rapid rate of
emptying is observed with large meal volume and initial
lag phase in emptying of small volume meals.

2.Composition of meal: The rate of gastric emptying for


various food materials in the following order
carbohydrates>proteins>fats.

3.Physical state: Liquid meal takes less time as


compared to solid meals.

4.GI pH: Gastric emptying is retarded at low stomach


pH and promoted at alkaline pH.
28-Mar-20 122
5.Exercise: Vigorous physical training retards gastric empting.

6.Body posture: Gastric emptying is favored while standing


and by lying on right side.

7. Emotional state: Stress and anxiety promotes GI motility,


where as depression retards it.

8.Drugs: That retards gastric emptying are antacids, anti


cholinergic, narcotic analgesics and tri cyclic antidepressants.
And metoclopramide, domperidone and cisapride stimulate
gastric emptying.

28-Mar-20 123
3. Intestinal transit
Defined as, the residence time of drug in small
intestine.
Delayed intestinal transit is desirable for:
1.Sustained release dosage forms,
2.Drug that only release in intestine ie ,enteric
coated formulations,
3.Drugs absorbed from specific sites in intestine,
eg; several B vitamins
28-Mar-20 124
4. GI pH
GI pH influence in several ways:
1.Disintegration: Disintegrating of some dosage forms is pH sensitive, enteric coated
tabs dissolve only in alkaline pH.
2.Dissolution: A large no. of drugs either weak acids or weak bases, their solubility is
greatly affected by GI pH.
-weakly acidic drugs dissolve rapidly in alkaline pH.
-basic drugs soluble in acidic pH.

3.Absorption: Depending upon drug pKa whether its an


acidic or basic drug the GI pH influences drug absorption.

4.stability of drug: GI pH influence the stability of drug.

Eg; erythromicin

28-Mar-20 125
5. Blood flow to git
GIT is extensively supplied by blood capillary, about
28% of cardiac output is supplied to GIT portion,
most drug reach the systemic circulation via
blood only.
Any factor which affects blood flow to GIT may also
affect absorption.

28-Mar-20 126
6. Disease state
Several disease state may influence the rate and extent of
drug absorption.
Three major classes of disease may influence bioavailability
of drug.

• GI diseases
• CVS disease
• HEPATIC disease

28-Mar-20 127
GI diseases
A. GI infections :
1.Celiac disease: (characterized by destruction of
villi and microvilli) abnormalities associated
with this disease are increase GI emptying
rate and GI permeability, alter intestinal drug
metabolism.
2.Crohn’s disease: alter gut transit time and
decreased gut surface area.
B. GI surgery:
Gastrectomy may cause drug dumping in
28-Mar-20 128
CVS diseases
In CVS diseases blood flow to GIT decrease,
causes decreased drug absorption.

Hepatic diseases
Disorders like hepatic cirrhosis influences bioavailability of
drugs which under goes first pass metabolism.

28-Mar-20 129
7. Gastro intestinal contents
1.Food- drug interaction: In general presence of food
either delay, reduce, increase or may not affect
absorption.
• Aspirin Delayed
• Penicillin's Decreased
• Griseofulvin Increased
• Methyldopa Unaffected
2.Interaction of drug with normal GI contents: GIT
contains no. of normal constituents such as mucin, bile
salts and enzymes, which influence the drug
absorption. Eg; Inhibitory action of bile on GI motility.
28-Mar-20 130
3.Drug-Drug interaction in the GIT:
Physico chemical drug- drug interaction:
Adsorption: Eg; anti diarrhial preparations
contains adsorbents like kaolin, prevents a
absorption of many drugs co-administered
with them.
Complexation: Eg; penicillin derivative with
ca-gluconate.
pH changes: Basic drugs changes gastric pH
28-Mar-20
Eg; tetracycline with antacids 131
8. First pass metabolism
Four primary systems which affect pre
systemic metabolism of a drugs.
1. Luminal enzymes.
2.Gut wall enzymes or mucosal
enzymes.
3. Bacterial enzymes.
4. Hepatic enzymes.

28-Mar-20 132
ABSORPTION OF DRUG FROM NON
PER OS EXTRAVASCULAR ROUTES
 NON PER OS Means other than oral routes which by passes the
GIT and reaches to systemic circulation.
 One of the major advantages of administering drugs by non-
invasive transmucosal (& transdermal) routes such as nasal,
buccal, rectal, etc. is that greater systemic availability is
attainable
ABSORPTION OF DRUG FROM NON
PER OS EXTRAVASCULAR ROUTES
• BUCCAL AND SUBLINGUAL
• RECTAL ADMINISTRATION
• TOPICAL ADMINISTRATION
• INTRAMUSCULAR ADMINISTRATION
• SUBCUATNEOUS ADMINISTRATION
• PULMONARY ADMINISTRATION
• INTRANASAL ADMINISTRATION
• INTRAOCULAR ADMINISTRATION
• VAGINAL ADMINISTRATION
BUCCAL AND SUBLINGUAL
 Buccal Route : The medicament is placed between cheek and the
gum.(Glyceryl trinitrate)
 Sublingual Route : The drug is placed under the tongue and allowed to
dissolve.(Ergotamine)
 Advantages :-
a) Rapid absorption
b) No first-pass hepatic metabolism
c) No degradation of drugs
 Factors:-
a) Lipophilicity of drugs
b) Salivary secretion
c) PH of the saliva
d) Binding to oral mucosa
e) Thickness of oral epithelium
RECTAL ADMINISTRATION
 The rectal route of administration is still an important route
for Children & Old Patients.
 The drug may be administered as solutions(microenemas) or
suppositories.
 Advantage :-
a) Absorption is more rapid

b) Bypasses presystemic hepatic metabolism

 Factors:-

1. Presence of faecal matter

2. pH of rectal fluid ( Around 8)

3. Drug Irritability

4. Surface area
TOPICAL ADMINISTRATION
 Skin is commonly employed as a site of drug administration
for local as well as systemic effect.
 Liquid dosage forms such as Liniments,Lotions, Sprays.
 Semisolids like Ointments,Creams,Pastes,Gels ,etc are
conventional drug forms for topical drug delivery
 Advantages:-
a) Protect drug from GI & from first pass metabolism
b) Increased patient compliance by reduced dosing frequency
c) Easy to terminate drug therapy by removing transdermal patch
 Factors:-
a) Skin condition
b) Composition of topical vehicle
c) Application procedure
d) External/environmental factors
INJECTIONS
 Intravenous(IV) Injection.
 Drug is directly goes into blood stream
 Intramuscular(IM) Injection.
 Absorption of drugs from I.M. sites is relatively rapid but
much slower than I.V. injection.

 Subcutaneous(SC) Injection.
 Absorption is slower than I.M. site due to poor perfusion
 Intraperitoneal(IP) Injection.
 I.P.route is rarely employed in human beings but most
widely used in laboratory animals
INJECTIONS
 Factors :-
a) Vascularity of injection site
b) Lipid solubility & Ionisation of drug
c) Molecular size of the drug
d) Volume of injection/Drug concentration
e) PH, composition & viscosity of injection
vehicle
PULMONARY ADMINISTRATION
 All drugs intended for systemic effect can be administered by inhalation
since the large surface area of the alveoli .

 Advantages:-
a) Rapid absorption just like exchange of gases between the blood and the
inspired air

b) Lipid-soluble drugs are rapidly absorbed by passive diffusion

c) Polar drugs absorbed by pore transport

 Factors:-
a) Particle size of drug
b) Properties of propeller such as vapour pressure,toxicity,solvent action

c) Effect of drugs and additives on mucous viscosity, mucocilliary clearence


INTRANASAL ADMINISTRATION
 The nasal route is becoming increasingly popular for systemic
delivery especially of some peptide and protein drugs

 Advantages:-
a) Rapid absorption due to rich vasculature and high permeability
b) Drugs from this route reaches the systemic circulation may cross
BBB
 FACTORS:-
a) Required high lipophilic drugs
b) Smaller molecular weight is required
c) pH of nasal secretion
d) Pathological condition
INTRAOCULAR ADMINISTRATION
 Topical application of drugs to the eyes is mainly meant for
local effects such as mydriasis, miosis, anaesthesia or
treatment of infections,gloucoma,etc.
 Advantages:-

• a) Lipophilic as well as Hydrophilic drugs are absorbed


• b) pH of lachrymal fluid influence
absorption of weak electrolytes
 Factors:-

a) Rate of blinking shows loss of drug

b) Viscosity of drug also affect on absorption


VAGINAL ADMINISTRATION
 Drugs meant for intravaginal application are
ganerally intended to act locally in the
treatment of bacterial or fungal infection or
prevent conception
 Advantages:-
a) Easy administration
b) Controlled delivery & termination of drug
action when desired, with this route
DRUG DISTRIBUTION
Introduction
After entry into systemic circulation, the drug is subjected to a
number of processes called as Disposition Processes that tend
to lower the plasma concentration. Two major disposition
processes are:

1. Distribution which involves reversible transfer of a drug


between compartments(blood and extravascular tissues).

2. Elimination which involves irreversible loss of drug from the


body. It comprises of biotransformation and excretion.

3/28/2020 MADHU 148


DISTRIBUTION
• COMPARTMENT ARE BLOOD AND
EXTRAVASCULAR TISSUES.
• PASSIVE DIFFUSION DEPENDENT
• CONCENTRATION AT SITE OF ABSORPTION
• IMPORTANT FOR ONSET, INTENSITY AND
DURATION OF ACTION.

3/28/2020 MADHU 149


STEPS IN DRUG DISTRIBUTION
PERMEATION OF FREE OR
BOUND DRUG PRESENT IN
BLOOD THROUGH
CAPILLARY WALL INTO ECF

PERMEATION OF DRUG
PRESENT IN ECF
THROUGH MEMBRANE
OF TISSUE CELLS INTO ICF

3/28/2020 MADHU 150


FACTORS AFFECTING DISTRIBUTION
OF DRUGS
Tissue Permeability of the Drug
a. Physiochemical Properties of the drug like Molecular
size, pKa and o/w Partition coefficient.
b. Physiological Barriers to Diffusion of Drugs.
Organ / Tissue Size and Perfusion Rate
Binding of Drugs to Tissue Components
a. Blood compartment
b. Extravascular Tissue Proteins
Miscellaneous Factors
Age, Pregnancy, Obesity, Diet, Disease states, and Drug
Interactions…

3/28/2020 MADHU 151


TISSUE PERMEABILITY OF DRUG
 Rate of Tissue Permeability, and
 Rate of Blood Perfusion.
 Physiochemical properties of drug
 Molecular size
 Degree of ionisation
 Stereochemical nature of drug
 Partition coefficient

 Physiological barriers to distribution of drugs


 Simple capillary endothelial barrier
 Simple Cell membrane barrier
 Blood brain barrier
 Blood CSF barrier
 Blood Placental Barrier
 Blood Testis barrier.
3/28/2020 MADHU 152
SIMPLE CAPILLARY ENDOTHELIAL BARRIER

Not a barrier to drugs.


Diffusion of All drugs Mol size < 600Da
(Ionised or unionized)
Only drugs bound to blood components are
restricted

3/28/2020 MADHU 153


SIMPLE CELL MEMBRANE BARRIER
• Entry of drug from ECF to ICF
• Depends upon permeability (lipophilicity)
• FREE DRUG
• IONISED AND UNIONISED DRUG
BLOOD • PROTEIN BOUND DRUG

• DRUGS SIZE <50 Da


• LIPOPHILIC DRUGS(50-600 Da)
• POLAR/IONISED DRUGS SIZE>50 Da
ECF • PROTEIN BOUND DRUGS CANNOT CROSS THE BARRIER

• BULK FLOW
• PASSIVE DIFFUSION
ICF • ACTIVE TRANSPORT

3/28/2020 MADHU 154


BLOOD BRAIN BARRIER
• The brain capillaries consist of endothelial cells
which are joined to one another by continous tight
intercellular junctions comprising Blood brain
barrier(BBB).
• LESS PERMEABLE TO WATER SOLUBLE DRUGS
• PRESENCE OF SPECIAL CELLS PERICYTES AND
ASTROCYTES.
• TRIGER AREA AND MEDIAN HYPOTHALMIC
EMINENCE-NO BBB
• NO PINOCYTOSIS
• DIRECT DIFFUSION FROM NOSE TO CNS
3/28/2020 MADHU 155
BLOOD BRAIN BARRIER

3/28/2020 MADHU 156


BLOOD BRAIN BARRIER

• TRANSPORTATION OF DRUGS
• Passive diffusion through the lipoidal barrier.
• Active transport of essential nutrients
• APPROACHES TO PROMOTE CROSSING THE
BBB
• USE OF PERMEATION ENHANCERS: DMSO
• OSMOTIC DISRUPTION OF BBB: MANNITOL IN
INTERNAL CAROTID ARTERY.
• USE OF DRUGS TO BRAIN: DIHYDROPYRIDINE REDOX
SYSTEM
3/28/2020 MADHU 157
BLOOD CSF BARRIER
• FORMED MAINLY BY CHROID PLEXUS OF THE
LATERAL, THIRD AND FOURTH VENTRICLE
• SIMILAR TO ECF OF BRAIN IN COMPOSITION
• CAPILLARY ENDOTHELIUM ALONG THE CHROID
PLEXUS HAVE OPEN JUNCTIONS FOR FREE FLOW OF
DRUG INTO ECF BETWEEN CAPILLARY WALL AND
CHOROIDAL CELLS.
• CHOROIDAL CELLS HAVE TIGHT JUNCTIONS
BETWEEN CHOROID CELLS SIMILAR TO BBB
• SINK CONDITION IS MAINATINED. CONC. IN BRAIN IS
ALWAYS HIGER THAN CSF.
3/28/2020 MADHU 158
BLOOD CSF BARRIER

3/28/2020 MADHU 159


BLOOD PLACENTAL BARRIER
• The maternal and the fetal blood vessels are separated by a
number of tissue layers made up of fetal tropoblast basement
membrane and endothelium which constitute placental
barrier.
• Thickness 25 -2 microns
• Diffusion drugs having
• Mol wt.<1000 Da
• Moderate to high lipid solubility
• Carrier mediated transport: essential nutrients
• Endocytosis: Immunoglobulins
• Better to restrict all drugs during pregnancy.
3/28/2020 MADHU 160
BLOOD PLACENTAL BARRIER

3/28/2020 MADHU 161


BLOOD TESTIS BARRIER
• Located at sertoli- sertoli cell junction
• Tight junction between sertoli cells
• Restricts passage of drugs to spermatocytes
and spermatids.

3/28/2020 MADHU 162


ORGAN/ TISSUE SIZE AND PERFUSION RATE

DISTRIBUTION DEPENDS UPON


PERMEABILITY
IONIC,POLAR, WATER SOLUBLE
RESTRICTION OF DIFFUSION BY HIGHLY SELECTIVE
PERMEABLE MEMBRANE.
PERFUSION
HIGHLY LIPOPHILIC
HIGHLY PERMEABLE MEMBRANES.

3/28/2020 MADHU 163


ORGAN TISSUE AND PERFUSION RATE

• Perfusion Rate is defined as the volume of blood that


flows per unit time per unit volume of the tissue.
• Greater the blood flow, faster the distribution
• Highly perfused tissues such as lungs, kidneys, liver,
heart and brain are rapidly equilibrated with lipid
soluble drugs.
• The extent to which a drug is distributed in a
particular tissue or organ depends upon the size of
the tissue i.e. tissue volume.

3/28/2020 MADHU 164


MISCELLANEOUS
• AGE
• PREGNANCY: Growth of uterus, placenta and fetus increase
volume for distribution of drugs.
• OBESTITY: In Obese persons, high adipose tissue content can
take up a large fraction of lipophilic drugs.
• DIET: A Diet high in fats will increase the free fatty acid levels
in circulation thereby affecting binding of acidic drugs such as
NSAIDS to Albumin.
• DISEASED STATE:
• Altered albumin or drug – binding protein conc.
• Altered or Reduced perfusion to organs /tissues
• Altered Tissue pH
3/28/2020 MADHU 165
PLASMA PROTEIN BINDING
Volume of Distribution

The Volume of distribution (VD), also known as Apparent


volume of distribution, is used to quantify the
distribution of a drug between plasma and the rest of the
body after oral or parenteral dosing.

It is called as Apparent Volume because all parts of the


body equilibrated with the drug do not have equal
concentration.
It is defined as the hypothetical volume of body fluid in
which drug is uniformly distributed or dissolved to
produce the observed blood concentration.

167
Redistribution
 Highly lipid soluble drugs when given by i.v. or by
inhalation initially get distributed to organs with high blood
flow, e.g. brain, heart, kidney etc.

 Later, less vascular but more bulky tissues (muscles,fat)


take up the drug and plasma concentration falls and drug is
withdrawn from these sites.

 If the site of action of the drug was in one of the highly


perfused organs, redistribution results in termination of the
drug action.
 Greater the lipid solubility of the drug, faster is its
redistribution.
168
The real volume of distribution has physiological meaning and
is related to the Body Water.

169
The volume of each of these compartments can be determined
by use of specific markers or tracers.
Physiological Fluid Compartments the Markers Used Approximate
volume (liters)
Plasma Evans Blue, 4
(totally bound to plasma albumin) Indocyanine
Green
Extracellular fluid-ECF Inulin, 14
(Easily penetrate capillary membrane , Raffinose,
rapidly distribute in ECF but do not cross Mannitol
cell membrane)

Total Body Water D2O, Antipyrine 42


(Equally distribute in all water
compartments of the body)

The intracellular fluid volume can be determined as the


difference between total body water and extracellular fluid.

170
Drugs which bind selectively to Plasma proteins e.g. Warfarin
have Apparent volume of distribution smaller than their Real
volume of distribution.

The Vd of such drugs lies between blood volume and total


body water i.e. b/w 6 to 42 liters.

Drugs which bind selectively to Extravascular Tissues e.g.


Chloroquine have Apparent volume of distribution larger than
their Real volume of distribution.

The Vd of such drugs is always greater than 42 liters.

171
PROTEIN BINDING OF DRUGS
• The phenomenon of complex formation of drug with
protein is k/w as drug protein binding.
• Protein binding is of two types:
– Intracellular binding : where a drug is bound to cell
proteins which may be a drug receptor to give a
pharmacological response called as primary receptors
– Extracellular binding: where a drug is bound to
extracellular proteins does not elicit a
pharmacological response called as secondary or
silent receptors
• pharmacologicaly/pharmacodynamically inert
• not metabolised
• nor excreted
• half life of drug increase.
MECHANISMS OF PROTEIN DRUG
BINDING
• Binding of drugs to protein is generally reversible
which involves weak chemical bonds
– hydrogen,
– hydrophobic,
– ionic
– and van der waal’s forces.
• Ireversible binding is rare and involve covalent
bonds.
– Carcinogenicity
– Tissue toxicity.
– eg chloroform & paracetamol metabolite results in
hepatotoxicity
BINDING COMPONENTS
• Binding of drug falls in two classes:
– Blood components
• Plasma proteins
– Human serum albumin
– α1- acid glycoprotein
– Lipoproteins
– α1 globulin
– α2 globulin
• Blood cells
– Haemoglobin
– Carbonic anhydrase
– Cell membrane
– extravascular tissue like tissue protein, fats, bones etc.
BLOOD COMPONENTS
• Following entry of drug into systemic circulation,
the drug interact with blood components like
plasma proteins, blood cells and heamoglobin.
• PP are present in abundant amounts and in large
variety.
• Drug – PPB is reversible
• Order of binding
– Albumin> α1- acid glycoprotein > lipoproteins
>globulins
Protein Molecular concentration Drug that bind
Weight (Da) (g/L)

Albumin 65,000 3.5–5.0 Large variety of drugs

α1- acid 44,000 0.04 – 0.1 Basic drug - propranolol, imipramine ,


glycoprotein and lidocaine .

Lipoproteins 200,000– .003-.007 Basic lipophilic drug


3,400,000 Eg- chlorpromazine
α1 globulin 59000 .003-.007 Steroid , thyroxine
Cynocobalamine

.015-.06 Vit. –A,D,E,K AND CUPRIC IONS


α2 globulin 13400

haemoglobin 64,500 11-16 Phenytoin, pentobarbitol,


phenothiazines.

176
HUMAN SERUM ALBUMIN
• Mol wt : 65,000
• Most abundant plasma protein (59% of total plasma)
• Conc. 3.5 to 5 %
• Large binding capacity
• Therapeutic doses of most drugs are much smaller and
their plasma drug conc. Do not normally reach equimolar
conc. with HSA.
• Bind to several components having varied structure.
• Have 4 binding sites
• Weak acids, neutral, weak bases can bind to HSA.
• A drug can bind to more than one site.
• Competitive binding.
BINDING OF DRUG TO HSA
• Site I: WARFARIN &
AZAPROPAZONE BINDING SITE
– eg. NSAIDS, Sulfonamides,
Phenytoin, Na valporate, bilirubin
• Site II: DIAZAPAM BINDING SITE
– benzodiazepenes, ibu, keto etc
• Site III: DIGITOXIN BINDING SITE
• Site IV: TAMOXIFEN BINDING
SITE
• Site I and II are responsible for
binding of most of drugs.
BINDING OF DRUG TO α1- Acid
Glycoprotein(α1AGP or AAG)
• Also called as orosomucoid
• Mol wt: 44,000
• Plasma conc. .04 to 0.1g%
• Binds to basic drugs like imipramine,
amitriptyline, nortriptyline, lidocaine,
propanolol, quinidine and disopyramide.
BINDING OF DRUG TO LIPOPROTEIN
• Bind to lipophilic drugs because of high lipophilic
contents.
• Plasma conc. Is less as compared to HSA and AAG
• Drug bind to lipoproteins by dissolving in lipid core
• Mol wt-2 to 34 lakhs
• Lipoprotein are classified into 4 categories depending
on density
– Chylomicron – less dense and largest in size
– VLDL-
– LDL- predominant in human
– HDL- most dense and smallest in size
BINDING OF DRUG TO LIPOPROTEIN
• Noncompetitive binding i.e. non-specific
binding site
• Binding is not dependent on drug
concentration
• Partitioning rather than binding
• Lipoprotein binding become significant when
HSA and AAG levels in plasma are decreased.
BINDING OF DRUG TO GLOBULIN
α1 globulin α2 globulin
(TRANSCORTIN or CBG)
bind to a number of steroidal
(CERULOPLASMIN ) bind
drug cortisone , prednisolone to Vit. A, D, E and K and
$ thyroxine , cynocobalamine cupric ions
γ- globulin
bind to specific
antigen

β1-globulin
β2-globulin
(TRANSFERRIN ) bind to
bind to carotinoid
ferrous ion
182
BINDING OF DRUG TO BLOOD CELLS
• 40% of Blood Comprises of blood cells
• Major component is RBC
• RBC is 500 times in diameter as compared to albumin
• Rate and extent of entry of into RBC is more for lipophilic
drugs (phenytoin).
• Hydrophillic drugs do not enter RBC (ampicillin)
• RBC comprises three components each of which can bind
to drugs
– Haemoglobin (mol wt. 64,500 but 7-8 times in conc than HSA)
– Carbonic anhydrase
– Cell membrane
Binding of drug to blood cells

184
TISSUE BINDING OF DRUGS
• Body tissues comprises 40% of the body
weight which is 100 times that of HSA.
• Importance
– Increases apparent volume of distribution of
drugs in contrast to PPB which decreases it.
– Tissue drug binding results in location of drugs at
a specific site in the body which increase shelf life
of drug in body.
– Tissue drug binding is irreversible. So cause tissue
toxicity.
Majority of drug bind to extravascular tissue-
the order of binding :-
liver > kidney > lung > muscle
liver – epoxide of number of halogenated
hydrocorban , paracetamol
lung – basic drug imipramine , chlorpramazine ,
antihistamine ,
kidney – metallothionin (a protein in kidney)
bind to heavy metal , lead, Hg , Cd ,
skin – chloroquine $ phenothizine (melanin is
present)
eye - chloroquine $ phenothizine (melanin is
present)
Hairs- arsenicals , chloroquine, $ PTZ bind to
hair shaft
Bone – tetracycline
186
COMPARISON
FACTOR AFFECTING DRUG-PB
• Drug related factors
– Physicochemical characteristic of drug
– Concentration of drug in the body
– Affinity of drug for a particular binding component
• Protein/Tissue related factors
– Physicochemical characteristic of the protein or binding agent
– Concentration of protein or binding component
– Num. Of binding site on the binding agent
• Drug interactions
– Competition b/n drugs for binding site (warfarin)
– Competition b/n drug and body constituents(kernicterus)
– Allosteric changes in protein molecule
• Patient related factor
– Age
– Intersubject variations
– Disease state
188
DRUG RELATED FACTOR
• PHYSICOCHEMICAL CHARACTERISTICS OF DRUG
 Protein binding is directly related to lipophilicity
lipophilicity = the extent of binding
• e.g. The slow absorption of cloxacilin in comparision to
ampicillin after i.m. Injection is attributes to its higher
lipophilicity, it bind 95% protein as comapred to 20% of
ampicillin.
• Highly lipophilic thiopental tend to localized in adipose tissue
• Anionic or acidic drug like . Penicillin , sulfonamide bind more
to HSA
• Cationic or basic drug like . Imipramine alprenolol bind to
AAG
189
DRUG RELATED FACTOR
CONCENTRATION OF DRUG IN THE BODY The extent of drug-
protein binding can change with both change in drug and
protein concentration
• The conc. of drug that bind HSA does not have much influence
as the therapeutic concentration of any drug is insufficient to
saturate it
Eg. therapeutic concentration of lidocaine can saturate AAG with
which it binding as the conc. Of AAG is much less in comparison
to that of HSA in blood
DRUG - PROTEIN / TISSUE AFFINITY
• Lidocaine have greater affinity for AAG than HSA
• Digoxin have greater affinity for protein of cardiac muscle than
skeleton muscles or plasma
190
PROTEIN / TISSUE RELATED FACTOR
PHYSICOCHEMICAL PROPERTY OF PROTEIN / BINDING
COMPONENT – lipoprotein or adipose tissue tend to bind
lipophilic drug by dissolving them to lipid core .
• The physiological pH determine the presence of anionic or
cationic group on the albumin molecule to bind a variety of
drug
CONCENTRATION OF PROTEIN / BINDING COMPONENT
• Mostly all drug bind to albumin b/c it present a higher
concentration than other protein
NUMBER OF BINDING SITES ON THE PROTEIN
• Albumin has a large number of binding site as compare to other
protein and is a high capacity binding component

191
 Several drug capable to binding at
more than one binding site Warferin binding si
e.g.- flucoxacillin , flurbiprofen ,
ketoprofen , tamoxifen and
dicoumarol bind to both primary and Site 1
secondary site of albumin Diazapam
Indomethacin is bind to three different Site 2 binding
site site
AAG is a protein with limited binding
Site 3
capacity b/c of it low conc. & Digitoxin
molecular Size . The AAG has only binding
one binding site for lidocaine , in site4
site
presence of HSA two binding site
have been reported due to direct
Tamoxifen
interaction b/w them
binding
Drug binding site onsite
HSA192
DRUG INTERACTIONS
COMPETITION B/W DRUG FOR BINDING SITE
(DISPLACEMENT INTERACTION )
• When two or more drug present to the same site , competition
b/w them for interaction with same binding site .
• If one of the drug (A) is bound to such a site , then administration of
the another drug (B) having high affinity for same binding site result
in displacement of drugs (A) from its binding site . This type of
interaction is known as displacement interaction .
• Where drug (A) here is called as the displaced drug and drug (B) as
the displacer .
• Eg. Phenylbutazone displace warfarin and sulfonamide from its
binding site

193
DRUG INTERACTIONS
COMPETITION B/W DRUG AND NORMAL BODY CONSTITUENT
• The free fatty acids interact with a number of drug that bind
primarily to HSA .
• When free fatty acid level is increase in several condition –
physiological (fasting) , pathologic (diabeties , myocardial infarction ,
alcohol abstinence) – the fatty acid which also bind to albumin
influence binding of several drug
binding – diazepam
- propanolol
binding - warfarin

Acidic drug like – Na.Salicilate , Na.Benzoate , sulfonamide displace


bilirubin from its albumin binding site. The free bilirubin not
conjugated by liver of neonates , crosses to BBB and precipitate
KERNICTERUS( degeneration of brain and mental retardation)
194
DRUG INTERACTIONS
ALLOSTERIC CHANGES
• The process involves alteration of protein
structure by the drug or its metabolite thereby
(allosteric effector) modifying its binding
capacity.
• Eg aspirin acetylates the lysine fraction of
albumin thus modifying its capacity to bind
NSAIDS. Phenybutazone ↑ and flufenamic
acid↓
PATIENT RELATED FACTOR
AGE – Modification in protein binding efficiency
• Neonate – albumin content is low in new born as result in
increase conc. of unbound drug that primarily bind to albumin
eg. Phenytoin , diazepam
• Young infants- eg digoxin- young infants suffering from CHF
are given a dose 4-6 times the adult dose on weight basis.
Reason- greater PB of digoxin in infants and large renal
clearance .
• Elderly -albumin content is lowered result in increase conc. of
unbound drug that primarily bind to albumin
 In old age- AAG level is increase thus decrease conc. of free
drug that bind to AAG
196
PATIENT RELATED FACTOR
DISEASE STATE
Disease Influence on Influence on protein
plasma protein drug binding

Renal failure Decrease binding of acidic drug ;


(uremia) albumin content neutral or basic drug are
unaffected

Hepatic failure Decrease binding of acidic drug ;


albumin synthesis binding of basic drug is normal or
reduced depending on AAG level.

Inflammatory Increase binding of basic drug ;


state AAG levels neutral and acidic drug unaffected
(trauma , burn,
infection )
197
SIGNIFICANCE OF PROTEIN/TISSUE BINDING OF
DRUGS
• ABSORPTION – the binding of absorbed drug to plasma proteins
decrease free drug conc. and disturb equilibrium . Thus sink
condition and conc. gradient are established which now act as the
driving force for further absorption
• SYSTEMIC SOLUBILITY OF DRUG water insoluble drugs ,
neutral endogenous macromolecules , like heparin , steroids , and
oil soluble vitamin are circulated and distributed to tissue by
binding especially to lipoprotein act as a VEHICLE for such drug
hydrophobic compound .
• DISTRIBUTION -The plasma protein-drug binding favors uniform
distribution of drug throughout the body by its buffer function . A
protein bound drug in particular does not cross the BBB, placental
barrier and the glomerulus. 198
SIGNIFICANCE OF PROTEIN/TISSUE
BINDING OF DRUGS
• TISSUE BINDING , APPARENT VOLUME OF
DISTRIBUTION AND DRUG STORAGE
 A drug that bind to blood component remains confined to
blood have small volume of distribution.
 Drug that show extra-vascular tissue binding have large
volume of distribution .
 the relationship b/w tissue drug binding and apparent
volume of distribution-
Vd = amount of drug in the body = X
plasma drug concentration C
• GREATER THE UNBOUND DRUG OR FREE CONCENTRATION
OF DRUG IN PLASMA, LARGER IS ITS Vd
SIGNIFICANCE OF PROTEIN/TISSUE
BINDING OF DRUGS
• ELIMINATION OF DRUG-
Protein binding reduced the elimination rate
due
Drug- protein complex due to it large size can
not the penetrate in to liver also prevents the
drug filtration from glomerulous………… half
life of drug will increase………..the drug remain
confined in body for long period of time.
SIGNIFICANCE …..
• DISPLACEMENT INTERACTION AND TOXICITY
Drug A Drug B

% DRUG BEFORE DISPLACEMENT


BOUND 99 90
FREE 1 10

% DRUG AFTER DISPLACEMENT


BOUND 98 89
FREE 2 11

% INCREASE IN FREE DRUG CONCENTRATION


100 10
Kernicterus – DI of bilirubin by NSAID’S drugs
Displacement of digoxine by qunidine
Displacement of warfarin by phenylbutazone
Interaction is significant if drug bind more than 95%
KINETICS OF PROTEIN DRUG BINDING
• The kinetics of reversible drug–protein binding for a protein
with one simple binding site can be described by the law of
mass action, as follows:

or ………………1
The law of mass action, an association constant, K a, can be expressed as the
ratio of the molar concentration of the products and the molar
concentration of the reactants. This equation assumes only one-binding
site per protein molecule
……………………….…2

Experimentally, both the free drug [D] and the protein-bound drug [PD], as
well as the total protein concentration [P] + [PD], may be determined.
To study the binding behavior of drugs, a determinable ratio (r )is defined, as
follows
202
moles of drug bound is [PD] and the total moles of protein is [P] +
[PD], this equation becomes
………………….3

Substituting the value of PD from equa. 2

…………4

This equation describes the simplest situation, in which 1 mole of drug binds to 1
mole of protein in a 1:1 complex. This case assumes only one independent
binding site for each molecule of drug. If there are n identical independent
binding sites per protein molecule, then the following is used:
………………..5
203
• In terms of K d, which is 1/K a, Equation 6
reduces to
……………….6

• Protein molecules are quite large compared to drug molecules


and may contain more than one type of binding site for the
drug. If there is more than one type of binding site and the
drug binds independently on each binding site with its own
association constant, then Equation 6 expands to

………….7
The values for the association constants Ka and the number of binding
sites n are obtained by various graphic methods.
204
1. Direct plot

It is made by plotting r
vresus (D)
At plateau,
r=n

2. Double reciprocal plot


The reciprocal of Equation 6 gives the following equation

205
• A graph of 1/r versus 1/[D] is called a
double reciprocal plot. The y
intercept is 1/n and the slope is
1/nKa . From this graph , the number
of binding sites may be determined
from the y intercept, and the
association constant may be
determined from the slope, if the
value for n is known.

3. Scatchardplot
is a rearrangement of Equation 6 The Scatchard plot spreads
the data to give a better line for the estimation of the binding
constants and binding sites. From Equation 6 , we obtain

206
– Biopharmaceutics and Pharmacokinetics A treatise by D.
M. Brahmankar Sunil B. Jaiswal. Page no. 220-244.
– Textbook of Biopharmaceutics and Pharmacokinetics by
Dr. Shobha Rani R. Hiremath. Page no. 85-89
– Biopharmaceutics and Clinical Pharmacokinetics An
Introduction by Robert E. Notari. Page no. 25-28 and 341-
342
– Clinical pharmacokinetics, a modern approach to
individualized drug therapy by Joseph Waztek.
– Applied Biopharmaceutics and pharmacokinetics by Leon
shargel.

207
THANKS

You might also like