Sie sind auf Seite 1von 106

Biopharmaceutics

Introduction to Biopharmaceutics
• Drugs obtained from plant, animal or mineral sources or
synthesized chemically, are rarely administered in their pure
chemical form.
• Often they are combined with a number of inert substances
(excipients & adjutants) and transformed into a convenient
dosage form that can be administered by a suitable route.
• Earlier, it was believed that the therapeutic response to a
drug is due to its intrinsic pharmacological activity.
• But today, it is very much understood that the dose-
response relationship obtained after drug administration by
different routes-for example, oral and parenteral, are not the
same.
• Variations are also observed when the same drug is
administered as different or similar dosage forms produced
by different manufacturers.
• It depends upon the physicochemical properties of the
excipients present in the dosage form, the method of
formulation & the manner of administration.
• Therefore a new and separate discipline called
biopharmaceutics has been developed to account for all such
factors that influence the therapeutic effectiveness of a drug.
• Biopharmaceutics is defined as the study of factors
influencing rate & amount of drug that reaches the systemic
circulation & the use of this information to optimize the
therapeutic efficacy of drug products.
• The process of movement of drug from its site of
administration to the systemic circulation is called as
absorption.
• The concentration of drug in plasma and hence the onset of
action, and the intensity and duration of response depend
upon the bioavailability of drug from its dosage form.
• Bioavailability is defined as the rate and extent (amount) of
drug absorption.
• Any alteration in the drug's bioavailability is reflected in
its pharmacological effects.
• Other processes that play a role in the therapeutic activity of
a drug are distribution and elimination. Together they are
known as drug disposition.
• The movement of drug between one compartment and the
other (generally blood and the extra vascular tissues) is
referred to as drug distribution.
• Since the site of action is usually located in the extra
vascular tissues, the onset, intensity and sometimes duration
of action depend upon the distribution behavior of the drug.
• The intensity and the duration of action depend largely upon
the effective concentration and the time period for which
this concentration is maintained at the site of action which
depend upon the elimination processes.
• Elimination is defined as the process that tends to remove
the drug from the body and terminate its action.
• Elimination occurs by two processes
1. Bio transformation (metabolism), which usually inactivates
the drug, and
2. Excretion which is responsible for the exit of
drug/metabolites from the body.
• In order to administer drugs optimally, knowledge is
needed not only of the mechanisms of drug absorption,
distribution, metabolism and excretion (ADME) but also
of the rate (kinetics) at which they occur i.e.
pharmacokinetics.
• Pharmacokinetics is defined as the study of time course of
drug ADME and their relationship with its therapeutic and
toxic effects of the drug.
• Pharmacokinetics is the kinetics of ADME.
• The use of pharmacokinetic principles in optimizing the
drug dosage to suit individual patient needs and achieving
maximum therapeutic utility is called as clinical
pharmacokinetics.
Phases of Drug Administration and Therapy

• Drug administration and therapy can be divided into four


phases or processes:
1. The Pharmaceutical Phase: It is concerned with
a) Physicochemical properties of the drug, and
b) Design and manufacture of an effective drug product for
administration by a suitable route.
2. The Pharmacokinetic Phase: It is concerned with the
ADME of drugs as extracted by the plasma drug
concentration-time profile and its relationship with the
dose, dosage form and dosing frequency and route of
administration. In short, it is the sum of
all the processes inflicted by the body on the drug.
• Pharmacokinetics is a study of what the body does to the
drug.
• Pharmacokinetics relates changes in concentration of drug
within the body with time after its administration.
3. The Pharmacodynamic Phase: It is concerned with the
biochemical and physiological effects of the drug and its
mechanism of action.
• It is characterized by the concentration of drug at the site
of action and its relation to the magnitude of effects
observed.
• Pharmacodynamic is a study of what the drug does to the
body.
• Pharmacodynamics relates response to concentration of
drug in the body.
4. The Therapeutic Phase: It is concerned with the
translation of pharmacological effect into clinical
benefit.
Drug + Excipients
Pharmaceutics

Drug not
Drug in Dosage form Released

Drug release & Drug


Dissolution decomposed,
Pharmacokinetics

metabolised,
bound or
Drug in Systematic excreted
Circulation

Extra Vascular tissues (at Metabolism


Site of action) & Excretion

Pharmacodynemics Pharmacological
Response

Therapeutics Therapeutic / Toxic


Effects
Role of Biopharmaceutics in Formulation development

• To achieve optimal therapy with a drug, the drug


product must be designed to deliver the active principle
at an optimal rate and amount, depending upon the
patient's needs.
• Knowledge of the factors affecting the bioavailability of
drug helps in designing such an optimum formulation
and saves many drugs that may be discarded as useless.
• On the other hand, rational use of the drug or the therapeutic
objective can only be achieved through a better
understanding of pharmacokinetics (in addition to
Pharmacodynamic of the drug), which helps in designing a
proper dosage regimen.
• A considerable experimentation is needed to arrive at the
balance between the desired therapeutic and the undesired
toxic effects in order to define an appropriate dosage
regimen.
• The knowledge & concepts of biopharmaceutics &
pharmacokinetics thus have an integral role in the design &
development of new drugs & their dosage forms &
improvement of therapeutic efficacy of existing drugs.
Drug Absorption
• It is the process by which a drug enters the bloodstream without being
chemically altered or the movement of a drug from its site of
application into the blood system.
• Intravascularly (intravenously and/or intra-arterially) administered
drug directly enters the systemic circulation and exerts its
pharmacological effects.
• After absorption, the effectiveness of a drug can be assessed by its
concentration at the site of action. However, it is difficult to measure a
drug concentration at the site of action. Instead, the concentration of
drug can be measured more easily and accurately in plasma. There is
always an existence of correlation between the plasma concentration
of a drug and the therapeutic response.
• Drug absorption is defined as the process of movement of unchanged
drug from the site of administration to systemic circulation.
Drug Absorption Patterns
• Based on the amount and the rate of absorption a drug may show any
of the three absorption patterns:
– Incomplete and slow absorption
– Complete but slow absorption
– Complete and rapid absorption
• A drug that is completely but slowly absorbed may fail to show
therapeutic response as the plasma concentration for desired effect is
never achieved. While rapidly absorbed drug attains the therapeutic
level easily to produce pharmacologic effect. Thus, both the rate and
extent of drug absorption are important.
• Complete and rapid absorption pattern has several advantages:
– Lesser susceptibility for degradation/interaction in GIT
– High drug blood level and rapid onset of action
– More uniform, greater and reproducible therapeutic response
Mechanisms of Drug Absorption
• The three broad categories of drug transport mechanisms
involved in absorption are':—
1. Transcellular /intracellular transport
2. Paracellular /intercellular transport
3. Vesicular transport
1. Transcellular/intracellular Transport is defined as the
passage of drugs across the GI epithelium. It is the most
common pathway for drug transport.
• The various Transcellular transport processes involved in
drug absorption are-
• Passive Transport Processes:
These transport processes do not require energy other than
that of molecular motion (Brownian motion) to pass through
the lipid bilayer. Passive transport processes can be further
classified into following types:
I. Passive diffusion
II. Pore transport
III. Ion-pair transport
IV. Facilitated- or mediated-diffusion
• Active Transport Processes:
These transport processes require energy to move drug
molecules from extracellular to intracellular. They are of
two types-
I. Primary active transport.
II. Secondary active transport
2. Paracellular / Intercellular Transport is defined as the
transport of drugs through the junctions between the GI
epithelial cells. This pathway is of minor importance in
drug absorption.
• The two paracellular transransport mechanisms involved in
drug absorption are-
I. Permeation through tight junctions of epithelial cells (insulin)
II. Persorption (occur in Lumen)
3. Vesicular or Corpuscular Transport (Endocytosis)
Like active transport, these are also energy dependent
processes but involve transport of substances within vesicles
into a cell.
• Since the mechanism involves transport across the cell
membrane, the process can also be classified as
Transcellular.
• Vesicular transport of drugs can be classed
two categories-
I. Pinocytosis
II. Phagocytosis
1. Passive Diffusion:
• Also called non-ionic diffusion. It is the major process for
absorption of more than “90%” of the drugs.
• The driving force for this process is the concentration
gradient.
• Concentration gradient is defined as the difference in the drug
concentration on both side of the membrane.
• Drug molecules move because of their kinetic energy. Since
no energy source is required, the process is called as passive
diffusion.
• During passive diffusion, the drug present in the aqueous
solution at the absorption site, partitions and dissolves in the
lipid material of the membrane and finally leaves it by
dissolving again in an aqueous medium.
• 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 the lower
concentration until equilibrium is attained and that the rate
of diffusion is directly proportional to the concentration
gradient across the membrane.
• It can be mathematically expressed by the following
equation:
dQ DAKm/w . (Cgit – C)
dt h
dQ/dt = rate of drug diffusion / rate of drug appearance in blood
D = diffusion coefficient of the drug through the membrane (area/time)
A = surface area of the absorbing membrane for drug diffusion(area)
Km/w = membrane water partition coefficient of the drug
Cgit – C = Concentration gradient and h = thickness of the membrane
Characteristics of Passive diffusion:
• The drug moves down the concentration gradient
indicating down-hill transport
• The rate of drug transfer is directly proportional to the
concentration gradient between GI fluid and the blood
compartment.
• Greater area and lesser thickness of the membrane
contribute to faster diffusion, thus the rate of absorption is
more rapid from the intestine than from the stomach.
• Equilibrium is attained when concentration gradient across
the membrane is zero.
• Greater the membrane water partition coefficient of drug,
faster the absorption.
• Since the membrane is lipoidal in nature, lipophilic drugs
diffuse at a faster rate than lipophobic drug.
• The rate of diffusion of unionized drugs is 3-4 times the
rate for ionized drugs.
• Diffusion is rapid when GI fluid volume is less, more
volume decrease diffusion by reducing the concentration
gradient, however this principle of GI fluid dilution is
useful in treating overdose or poisoning of drug.
• Passive diffusion process is energy independent and
nonsaturable but dependent on square root of the molecular
size of the drug. Drugs with molecular weights between
100-400 Daltons are effectively absorbed by passive
diffusion.
Kinetics of Passive diffusion:
• Initially, when the drug is ingested, CGIT >> C, therefore
greater concentration gradient exists for drug absorption.
After equilibrium, the drug diffusion would stop as per
fick’s law, but this is not the case for invivo. Once the
passively absorbed drug enters the blood, it is rapidly
swept away and distributed in to a large volume of body
fluids. Hence, the concentration of drug at absorption site
(CGIT) is maintained greater than the concentration of drug
in plasma. Such a condition is called as sink condition for
drug absorption.
• Under usual conditions of absorption, D, A, Km/w and h are
constant and can be replaced by a combined constant, P,
called permeability coefficient.
• Permeability refers to the ease with which a drug can
penetrate or diffuse through a membrane.
• Moreover, due to sink conditions, the concentration of
drug in plasma (C) is very small in comparison to the
concentration of drug in GIT (CGIT); the equation can be
simplified to
dQ/dt = P x CGIT
• This is the equation for first order process. Thus the
passive diffusion follows first order kinetics.
2. Pore Transport:
• It is also called as convective transport, bulk flow or
filtration.
• Mechanism is responsible for transport of molecules into
the cell through the protein channels present in the cell
membrane.
• Following are the characteristics of pore transport:
1. The driving force is constituted by the hydrostatic pressure
or the osmotic differences across the membrane due to
which bulk flow of water along with small solid molecules
occurs through such aqueous channels. Water flux that
promotes such port is called as solvent drag.
2. The process is important in the absorption of low
molecular weight (less than 100), low molecular size
(smaller than diameter of the pore) and generally water-
soluble drugs through narrow, aqueous-filled channels or
pores in the membrane structure – for example, urea, water
& sugars.
3. Chain-like or linear compounds of molecular weight up to
400 Daltons can be absorbed by filtration.

• Drug permeation through water-filled channels is of


particular importance in renal excretion, removal of drug
from the cerebrospinal fluid and entry of drugs into the
liver.
3. Ion pair transport:
• This mechanism explains the absorption of drugs that ionize
under all pH conditions. E.g. quaternary ammonium
compounds and sulfonic acids.
• Such ionic drugs penetrate the membrane by forming
reversible neutral complexes with an oppositely charged
ions or endogenous ions of GIT like, mucin.
• Such neutral complexes have both the required lipophilicity
and aqueous solubility and diffuse more easily across the
membrane by passive diffusion.
• Ion paring of propranolol with oleic acid and quinine with
hexysalicylate are the demonstrated example to facilitate
drug absorption by this mechanism.
4. Carrier-Mediated Transport:
• Some polar drugs cross the membrane more readily than
their crossing capacity predicted from their concentration
gradient and partition coefficient values.
• This is because of the specialized transport mechanisms
called Carrier mediated transport (CMT) without which
many essential water-soluble nutrients like monosaccharide,
amino acids and vitamins will be poorly absorbed.
• The carrier is membrane enzyme/protein that binds in
reversible/non covalent manner with the molecules to be
transported.
• This carrier-molecule complex passes across the membrane
to the other side where it dissociates and discharges the
molecule.
Characteristics of carrier mediated transport:
• The process is structure specific i.e. carriers have special
affinity for only specific chemical structure for transport.
Generally, the carriers have special affinity for essential
nutrients.
• As the system is structure specific, drug having structure
similar to essential nutrients, called as False nutrients, are
absorbed by the same carrier system. Thus, it serves for the
absorption of several antineoplastic drugs like 5-
fluorouracil, 5-bromouracil which are false nutrients.
• Since the numbers of carriers are limited, the system is
capacity limited i.e. the system becomes saturated and
approaches a platue (Constant) at higher concentration.
• In case of passive diffusion the rate of absorption increases
linearly with concentration. While in case of CMT,
• drug absorption increases linearly with concentration until
the carriers become saturated after which it becomes
curvilinear and approaches a constant value at higher doses.
• Such process can be well described by mixed order kinetics
(first order at sub-saturation concentration and apparent zero
order at and above saturation concentration). It is also called
as Michaelis-Menten, saturation or non linear kinetics.
• Bioavailability decreases with increasing dose, due to
limited carriers, which are saturated at higher doses. Hence
it is irrational to administer large single oral dose of such
drugs like vitamin B1, B2 and B12.
• As the members of carriers are limited, the system is subject
to competition between agents having structure similarity.
• Carrier mediated absorption generally takes place from
specific sites of the GIT which are rich in number of
carriers. The area of the GIT in which the carriers are most
dense is called as absorption window. Drugs absorbed
through such absorption window are poor candidates for
conventional controlled release formulations but good
candidates for site specific drug delivery.
5. Facilitated Diffusion:
• It is a diffusion process facilitated by the carrier.
• It requires the help of carrier (usually protein) hence also
called carrier mediated transport.
• It operates down the concentration gradient hence called
downhill transport.
• It occurs at a much faster rate than passive diffusion.
• The driving force is concentration gradient.
• It is energy independent, hence not inhibited by metabolic
poisons that interfere with energy production.
• It has limited importance in the absorption of drugs.
• This process is important for entry of glucose into RBCs
and intestinal absorption of vitamin B1, B2 and B12.
• Sometimes extra component may be required in addition to
the carrier to facilitate the diffusion for example vitamin
B12 is absorbed by intrinsic factor (IF), a glycoprotein
produced by the gastric parietal cells. IF forms complex
with vitamin B12 which is then transported across the
intestinal membrane by a carrier system.
6. Active Transport:
• This requires the help of carrier hence called carrier
mediated transport.
• It is more important than facilitated diffusion in the
absorption of drugs and nutrients.
• The drug is transported from a region of lower
concentration to higher concentration i.e. against the
concentration gradient. Hence it is up hill transport.
• Energy is required in the work done by the carrier.
Therefore, it is energy dependent process which can be
inhibited by metabolic poisons that interfere with energy
production like fluorides, cyanides, dinitrophenol and lack
of oxygen.
• Endogenous substances transported actively are ions like
Na+, K+, Ca+2, iron, certain amino acids and vitamins like
ascorbic acid, niacin, pyridoxine.
• Drugs having structure similarity with endogenous
substances are absorbed by active transport.
• Drugs having structure similarity may undergo competitive
inhibition for absorption. E.g. impaired absorption of
levodopa when taken with meal rich in protein.
• Active transport is also important in renal and biliary
excretion of many drugs and their metabolites.
• There are two types of active transport systems:
– Ion pumps
– Co-transport.
Ion pumps:
• Ion pumps also called electrogenic pumps are special
proteins which actively transport ions resulting in an ionic
and voltage gradient. This gradient is a form of stored
energy used to fuel other processes.

Co-transport:
• It is the process of coupled passage of two materials across
a membrane. One type of material enters the cell and open
a door for other type of material. For example hydrogen
opens a door for intracellular entry of sucrose.
7. Carrier mediated intestinal transport:
• Various carrier mediated systems are present at the intestinal
brush border and basolateral membrane for the absorption
of specific ions and nutrient essential for the body.
• Many drugs are absorbed by these carriers because of the
structural similarity to natural substances.
• A transmembrane protein, P-glycoprotein has been
identified in the intestine which reduces apparent intestinal
epithelial cell permeability from intestinal lumen to blood
for various lipophilic or cytotoxic drugs.
• There are many transporters present in the intestine that help
in the absorption of drugs. For example, many oral
cephalosporins are absorbed through the oligopeptide
transporter.
8. Endocytosis:
• It is the process of engulfing particles or dissolved materials
by the cell.
• It involves engulfing extracellular materials within a
segment of the cell membrane to form a saccule or a vesicle
which is then pinched-off intracellularly.
• It is also called as corpuscular or vesicular transport. It is a
minor transport mechanism.
• Exocytosis is the process of moving macromolecules out of
a cell. An example of exocytosis is the transport of protein
such as insulin producing cells of the pancreas into the
extracellular space.
• Sometimes, an endocytic vesicle is transported from one
extracellular compartment to another, which is called
transcytosis.
• This process is responsible for the cellular uptake of
macromolecular nutrients like fats and starch, oil soluble
vitamins like A, D, E and K, water soluble vitamin like B12
and drugs such as insulin.
• The drug is directly absorbed into the lymphatic circulation
by this process there by bypassing first-pass hepatic
metabolism.
• Orally administered sabin polio vaccine, large protein
molecule and the botulism toxin (that causes food
poisoning) are thought to be absorbed by this process.
• This is the only process for which aqueous solution of drug
is not a prerequisite for absorption.
• Endocytosis includes two types of processes:
1. Phagocytosis (cell eating): is an uptake of larger
particles or macromolecules generally by macrophages.
2. Pinocytosis (cell drinking): is an uptake of small solute
or fluid.
3. Receptor mediated endocytosis: is the endocytosis
process in which, vesicle is formed after binding of
molecule with the receptors present on membrane.
7. Ionic diffusion:
• It is also called as electrochemical diffusion.
• The charge on membrane influences the permeation of
drugs.
• Molecular forms of drugs are unaffected by the membrane
charge and permeate faster than ionic forms.
• At a given pH, the rate of permeation is in the order of
unionized > anions > cations.
• Membrane is positively charged hence cations having high
kinetic energy can only permeate the ionic barrier.
• The ions are moving downhill the electrical gradient.
• Once inside the membrane, cations are attracted by
negatively charges intracellular membrane and the process
is continued until equilibrium, like passive diffusion.
Biopharmaceutical Consideration in DFD
• The drug product must deliver the active drug at an optimum rate and
amount to achieve the desired therapeutic objective.
• By proper biopharmaceutic design, the rate and extent of drug
absorption can be varied from rapid and complete absorption to slow
and sustained absorption depending upon desired therapeutic
objective.
• The sequence of events that occur after oral administration of a solid
dosage form such as tablet or capsule until it absorption into systemic
circulation consists of four steps:
– Disintegration of the dosage form
– Deaggregation and subsequent release of the drug
– Dissolution of the drug in aqueous fluids at the absorption site.
– Movement of dissolved drug through the GI membrane into the systemic
circulation.
• Unless the drug goes into solution, it cannot be absorbed into the
systemic circulation.
• In a series of kinetic or rate processes, the rate at which the drug
reaches the systemic circulation is determined by the slowest of the
various steps involved in the sequence. Such a step is called as the rate
determining step or rate limiting step.
• The rate and extent of drug absorption from its dosage form can be
influenced by a number of factors in all these steps.
Factor affecting GI Absorption of a Drug
A. Pharmaceutical Factors:
I. Physicochemical Properties of Drug Substances
1. Drug solubility & Dissolution rate
2. Particle size & effective surface area
3. Polymorphism & amorphism
4. Pseudopolymorphism
5. Salt form of the drug
6. Lipophilicity of the drug
7. pKa of the drug & gastrointestinal pH
8. Drug stability
II. Dosage form Characteristics & Pharmaceutical Ingredients
(Pharmaco-technical Factors)
1. Disintegration time (Tablets/capsules)
2. Dissolution time
3. Manufacturing variables
4. Pharmaceutical ingredients (excipients/adjuants)
5. Nature & Type of dosage form
6. Product age & Storage condition
B. Patient Related Factors:
1. Age
2. Gastric emptying time
3. Intestinal transit time
4. Gastrointestinal pH
5. Disease states
6. Blood flow through the GIT
7. Gastrointestinal Contents:
a. Other drugs
b. Food
c. Fluids
d. Other normal GI contents
8. Presystemic metabolism by:
a. Luminal enzymes
b. Gut wall enzymes
c. Bacterial enzymes
d. Hepatic enzymes
Factor affecting GI Absorption of a Drug
A. Pharmaceutical Factors:
I. Physicochemical Properties of Drug Substances
1. Drug solubility & Dissolution rate
2. Particle size & effective surface area
3. Polymorphism & amorphism
4. Pseudopolymorphism
5. Salt form of the drug
6. Lipophilicity of the drug
7. pKa of the drug & gastrointestinal pH
8. Drug stability
Drug Solubility & Dissolution Rate:
• Except in case of controlled release formulations,
disintegration and deaggregation occur rapidly from a well-
formulated dosage form. Thus, the two critical slower rate-
determining processes in the absorption of orally
administered drugs are:
1. Rate of drug dissolution
2. Rate of drug permeation through the biomembrane
• Dissolution is the RDS for hydrophobic poorly aqueous
soluble drugs like griseofulvin and spironolactone;
absorption of such drugs is often said to be dissolution rate-
limited.
• If the drug is hydrophilic with high aqueous solubility-for
example, cromolyn sodium or neomycin, then dissolution is
rapid and RDS of such drugs is rate of permeation through
the biomembrane. Absorption of such drugs is said to be
permeation rate-limited or transmembrane rate-limited.
• Presence of drug in aqueous solution is an important
requirement for the absorption of a drug by all mechanism
except endocytosis. This depends on the drug’s aqueous
solubility and its dissolution rate.
• Absolute or intrinsic solubility: is defined as the maximum
amount of solute dissolved in a given solvent under standard
conditions of temperature, pressure and pH. It is a static
(unchange) property.
• Dissolution rate: is defined as the amount of solid substance
that goes into solution per unit time under standard
conditions of temperature, pH and solvent composition and
constant solid surface area. It is a dynamic (Change
constantly) process.
• Drugs with poor aqueous solubility show dissolution
problems. However, there are well known examples of
drugs which in spite of low aqueous solubility have
sufficient oral bioavailability.
• Reasons for this may be the rapid rate of dissolution despite
low intrinsic solubility and due to small dose the GI transit
time is sufficient for adequate dissolution and absorption to
occur.
• Thus, the dynamic process of drug dissolution is better
related to drug absorption and bioavailability than absolute
solubility.
Theories for drug dissolution:
• The process for drug dissolution involves mass transfer
from the solid surface to the liquid phase. The three
important amongst the several theories are:
– Diffusion layer model or Film theory
– Danckwert’s model or surface renewal theory
– Interfacial barrier or double barrier model or limited salvation
theory
Diffusion layer model/ Film theory:
• It is the simplest and most common theory for dissolution.
This theory states that the process of dissolution consists of
two consecutive steps:
1. Solution of a solid to form a thin film or a layer at the
solid/liquid interface called as the stagnant film or
diffusion layer, which is saturated with the drug. This step
is usually rapid.
2. Diffusion of solute from a stagnant layer to the bulk of
solution. This step is slower and is therefore the RDS in
drug dissolution.
• The dissolution rate based on this theory can be assessed by
Noyes-Whitney equation, which is derived using following
assumptions:
1. The dissolving drug particle are spherical in shape and
equal in size
2. The dissolution process is controlled by diffusion of
molecules or ions.
3. There is no chemical interaction between the dissolving
drug and the components of the dissolving fluid.
4. The thickness of the diffusion layer and the saturation
solubility of drug in diffusion layer are constant
irrespective of the particle size.
• According to Noyes-whitney, the rate of dissolution is given
by:
• Bruner incorporated Fick’s first law of diffusion and
modified Noyes-Whitney equation to

• Where,
– Kw/o = water/oil partition coefficient of the drug-intrinsic
dissolution rate constant
– V = volume of dissolution medium
• Based on film theory, the effect of different parameters on
dissolution rate is as follows:
• As the D, A, Kw/o, Cs – Cb value increases, dissolution
increases and as the h value increases dissolution decreases.
• Dissolution follows first order process with the
concentration gradient as a driving force under non sink
conditions (in vitro) and zero order kinetics under sink
conditions (in vivo). To obtain good IVIVC in dissolution
rate, the in vitro dissolution must be carried out under sink
conditions.
• These invitro sink conditions can be achieved by:
– Bathing the dissolving solid in fresh solvent from time to time
– Increasing the volume of dissolution fluid
– Adding water miscible solvent to the dissolution fluid
– By adding selected adsorbents to remove the dissolved drug
Limitations of film theory:
• Assumption of constant surface area during dissolution by
this theory is practically not possible for dissolving
particles.
• To account for the particle size decrease and change in
surface area during dissolution, Hixon and Crowell’s cube
root law of dissolution is used.
Danckwert’s Model/ Surface renewal theory:
• Danckwert never agreed for the existence of a stagnant layer
and suggested that turbulence in the dissolution medium
exists at the solid/liquid interface.
• As a result, the agitating fluid consisting of macroscopic
packets of fluid that reach the solid/liquid interface in a
random fashion and absorb the solute by diffusion and carry
it to the bulk of the solution.
• Such solute containing packets are continuously replaced
with new packets of fresh solvent due to which the drug
concentration at the solid/liquid interface never reaches to
Cs and has a lower intermediate limiting value (Ci).
• Since the solvent packets are exposed to new solid surface
each time, the theory is called surface renewal theory.
• The model is expressed by equation:

• Where,
• M – mass of solid dissolved
• γ – interfacial tension or rate of surface renewal
Interfacial Barrier model/ Limited solvation theory:
• The diffusion layer model and the Danckwert’s model are
based on two assumptions:
– The rate determining step that controls dissolution is the mass
transport (diffusion)
– Solid solution equilibrium is achieved at the solid/ liquid interface.
• According to the interfacial barrier model, an intermediate
concentration can exist at the interface as a result of
salvation mechanism and is a function of solubility rather
than diffusion.
• Such concept is given by the equation:
dm/dt = Ki (Cs – Cb)
• Where Ki : effective interfacial transport rate constant
• Of the various factors, the factor of prime importance is
drug solubility. Almost every factor that affects dissolution
rate influences the drug solubility in one or other way.
• Dissolution rate is directly related to drug solubility.
• The equation useful to predict the dissolution rate of a drug
from its solubility is :
• Dissolution rate (R) = dc/dt = 2.24Cs
• The drug should have a minimum aqueous solubility of 1%
to avoid bioavailability problems.
Particle Size and Effective Surface Area of the Drug:
• Particle size and surface area of a solid drug are inversely
related to each other.
• Smaller the drug particle, greater the surface area.
• Two types of surface area of interest can be define:
1. Absolute surface area which is the total area of solid
surface of any particle,
2. Effective surface area which is the area of solid surface
exposed to the dissolution medium. The dissolution rate is
directly related to this effective surface area.
• The first approach to increase the dissolution of the drug is
to reduce the particle size of drug to micron level by
micronization.
• The effective surface area of nonhydrophobic drugs can be
increased by micronization. The surface area of such small
particles has higher energy than the bulk of the solid,
resulting in an increased interaction with the solvent. Thus,
micronization of poorly aqueous soluble drug results in
superior dissolution rates in comparison to simple milled
form of these drugs.
• However, in case of hydrophobic drugs, micronization
results in a decrease in the effective surface area of such
drugs and thus, a fall in the dissolution rate. The reasons for
this outcome are:
1. Hydrophobic drugs adsorb air onto their surface which
inhibits their wettability due to which the powders float on
the dissolution medium.
2. Due to high surface free energy, particles tend to form
aggregate which either float on the surface or settle at the
bottom of the dissolution medium.
3. Extreme size reduction in dry state imparts surface charges
that may prevent wetting.
• The absolute surface area of hydrophobic drugs can be
converted to their effective surface area by using wetting
agents and by adding hydrophilic diluents.
• The second approach to increase the dissolution is increase
in its solubility by reducing the particle size to a submicron
level by use of following specialized techniques:
– Formation of molecular dispersion: where the drug is trapped in
the lattice of a hydrophilic agent such as cyclodextrins.
– Formation of solid dispersion: where the drug is dispersed in a
soluble carrier such as PVP or PEG.
• Particle size reduction and increase in the surface area and
dissolution rate is not always advisable when the drugs are
unstable and degrade in solution form, produce undesirable
effects or when a sustained effect is desired.
Polymorphism and Amorphism:
• Depending upon the internal structure, a solid can exist
either in a crystalline or amorphous form.
• When a substance exists in more than one crystalline form,
the different forms are designated as polymorphs and the
phenomenon as polymorphism.
• Polymorphs are of two types:
1. Enantiotropic polymorph is the one which can be
reversibly changed into another form by altering the
temperature or pressure e.g. sulphur.
2. Monotropic polymorph is the one which is unstable at all
temperatures and pressures. e.g. glyceryl stearate.
• The polymorphs differ from each other with respect to their
physical properties such as solubility, melting point, density,
hardness and compressibility.
• They can be prepared by crystallizing the drug from
different solvents under diverse conditions.
• The existence of the polymorphs can be determined by
using techniques such as optical crystallography, X-ray
diffraction, differential scanning calorimetry etc.
• Depending on their relative stability, one of the several
polymorphic forms will be physically more stable than the
others.
• Such a stable polymorph represents the lowest energy state,
has highest melting point and least aqueous solubility.
• The remaining polymorphs are called as metastable forms
which represent the higher energy state, have lower melting
points and higher aqueous solubilities.
• Because of their higher energy state, the metastable forms
have a thermodynamic tendency to convert to the stable form.
A metastable form cannot be called unstable because if it is
kept dry, it will remain stable for years.
• Since the metastable forms have greater aqueous solubility,
they show better bioavailability and are therefore preferred in
formulation. Example: Chloramphenicol palmitate exists in
• A, B and C in which B shows best bioavailability while form
A is biologically inactive.
• The transformation of metastable form to stable form can be
inhibited by dehydrating the molecule environment or by
adding viscosity building macromolecules such as PVP, CMC,
pectin or gelatin.
• Some drugs exist in amorphous form and have highest energy
state and can be considered as super cooled liquids. They have
greater aqueous solubility than crystalline forms.
• The order of dissolution of different solid forms of drugs is:
amorphous > metastable crystalline > stable crystalline.
Hydrates/Solvates (pseudopolymorphism):
• The crystalline form of a drug can either be a polymorph or
molecular adduct or both.
• The stoichiometric type of adducts where the solvent
molecules are incorporated in the crystal lattice of the solid
are called as the solvates, and the trapped solvent as solvent
of crystallization.
• The solvates can exist in different crystalline forms called as
pseudopolymorphs. This phenomenon is called as
pseudopolymorphism.
• When the solvent in association with the drug is water, the
solvate is known as a hydrate.
Salt form of the Drug:
• Most drugs are either weak acids or weak bases. Easiest way to
enhance the solubility & dissolution rate of such drugs is to
convert them into their salt forms.
• Weak acids such as barbiturates & sulphonamides are converted
to strong base salt such as sodium & potassium. Weak base like
alkaloidal drugs are converted to strong acid salts such as
hydrochoride or sulphate.
• The pH of the diffusion layer of the salt form of a weak acid will
be higher than that observable with the free acid form of the
drug. This higher pH favors the dissolution of weak acids.
• In case of salts of weak bases, the pH of the diffusion layer will
be lower in comparison to that found with the free base form of
the drug. Consequently, the solubility of a basic drug at lower
pH is enhanced.
Drug pKa, GI pH and Lipophilicity of the drug (ph-partition hypothesis)
• The pH partition theory explains process of drug absorption
from the GIT and its distribution across all biological
membranes.
• The theory states that for drug compounds of molecular
weight greater than 100, which are transported across the
biological membrane by passive diffusion, the process of
absorption is governed by:
1. The dissociation constant (pKa) of the drug
2. The lipid solubility of the unionized drug
3. The pH at the absorption site
Drug pKa and Gastrointestinal pH:
• Ionization of drug is a function of drug pKa and pH of the
fluid at the absorption site.
• Dissociation constant of both acidic and basic drugs are
expressed by pKa values. Lower the pKa value for acid,
stronger is the acid and higher the pKa value for base,
stronger is the base.
• From the pKa values and pH at absorption site, the
percentage of drug ionized at this pH can be determined by
Henderson Hasselbach equation.
• For weak acids: pH = pKa + log[Ionized / unionized]
• For weak bases: pH = pKa + log[Unionized / Ionized]
Lipophilicity of drug:
• Even if the drug exists in the unionized form, it will be
poorly absorbed if it has poor lipid solubility.
• For optimum absorption, a drug should have sufficient
aqueous solubility to dissolve in the fluids at the absorption
site and lipid solubility high enough to cross the
biomembrane. In short, a perfect HLB should be there in the
structure of the drug for optimum bioavailability.
• Lipid solubility of drug is determined from its oil/water
partition coefficient value. In general, the octanol/pH 7.4
buffer partition coefficient value of 1-2 is sufficient for
absorption of drug, if it is present in unionized form.
• Thus, to enhance the bioavailability, in addition to the
dissolution rate, permeability is also important which can be
promoted by modification of chemical structure of the drug.
Limitation of pH-partition Hypothesis:
• Presence of virtual (microclimate) pH
• Absorption of ionized drugs
• Influence of GI surface area and residence time of drug
• Presence of aqueous unstirred diffusion layer
Drug stability:
• A drug for oral use may destabilize either during its shelf-
life or in the GIT.
• Two major stability problems resulting in poor
bioavailability of an orally administered drug are-
1. Degradation of the drug into inactive form, and
2. Interaction with one or more different components either
of the dosage form or those present in the GIT to form a
complex that is poorly soluble or is unabsorbable.
Factor affecting GI Absorption of a Drug
II. Dosage form Characteristics & Pharmaceutical Ingredients
(Pharmaco-technical Factors)
1. Disintegration time (Tablets/capsules)
2. Dissolution time
3. Manufacturing variables
4. Pharmaceutical ingredients (excipients/adjuants)
5. Nature & Type of dosage form
6. Product age & Storage condition
Disintegration time:
• Disintegration time (DT) is of particular importance in case
of solid dosage forms like tablets and capsules.
• In vitro disintegration test does not give guarantee of drug’s
bioavailability because if the disintegrated drug particles do
not dissolve, absorption is not possible.
• However, if a solid dosage form does not conform to the
DT, it shows bioavailability problems because the
subsequent process of dissolution will be much slower and
absorption may be insufficient.
• Rapid disintegration is an important in the therapeutic
success of a solid dosage form.
• DT of a tablet is directly related to the amount of a binder
present and the compression force of a tablet (Hardness).
Manufacturing / Processing variables:
• Drug dissolution is the single most important factor in the
absorption of drugs, especially from widely used
conventional solid dosage form, tablets and capsules.
• The dosage form related factors that influence dissolution
and hence absorption of a drug from such formulations are:
1. Excipients and
2. Manufacturing processes
• The influence of excipients such as binders, lubricants,
disintegrants, etc. on drug dissolution. Several
manufacturing processes influence drug dissolution from
solid dosage form. Processes of such importance in the
manufacture of tablets are:
1. Method of granulation
2. Compression force
Pharmaceutical ingredients (excipients/adjuants)
• A drug is rarely administered in its original form.
• Almost always, a convenient dosage form to be
administered by a suitable route is prepared. Such a
formulation contains a number of excipients. Excipients are
added to ensure acceptability, physicochemical stability
during the shelf-life, uniformity of compression and
optimum bioavailability and functionality of the drug
product.
• Despite their inertness and utility in the dosage form,
excipients can influence the absorption of drugs. More the
number of excipients in a dosage form, more complex it is
and greater the potential for absorption and bioavailability
problems.
Nature & Type of dosage form
• A part from the proper selection of drug, clinical success
often depends to a great extent on the proper selection of
dosage form of that drug.
• For a given drug 2 to 5 fold or perhaps more difference
could be observed in the oral bioavailability of a drug
depending upon the nature and type of dosage form.
• Such a difference is due to the relative rate at which a
particular dosage form releases the drug to the biological
fluid and the membrane.
• As a general rule, the bioavailability of a drug from various
dosage forms decreases in following order:
Solutions > Emulsions > Suspensions > Capsules > Tablets > Coated
Tablets > enteric coated tablets > Sustain release Tablets
Product Age & Storage Conditions:
• A number of changes, especially in the physicochemical
properties of a drug in dosage form, can result due to aging
and alteration in storage conditions which can adversely
affect bioavailability .
• Changes that occur during the shelf life of a dosage form are
affected mainly by large variations in temperature and
humidity. In one of the studies conducted on prednisone
tablets containing lactose as the filler, high temperature and
high humidity resulted in harder tablets that disintegrated
and dissolved slowly.
• Aging and improper storage may lead to:
– Precipitation of drug due to altered solubility because of
conversion of metastable polymorph to poorly soluble stable
polymorph in solution.
– Change in particle size distribution due to crystal growth in
suspension due to fluctuations in temperature during storage
– In tablets, decreased disintegration / dissolution due to binder that
harden on storage or increase in the same due to softening of
binder during storage may be observed.
B. Patient Related Factors:
1. Age
2. Gastric emptying time
3. Intestinal transit time
4. Gastrointestinal pH
5. Disease states
6. Blood flow through the GIT
7. Gastrointestinal Contents:
a. Other drugs
b. Food
c. Fluids
d. Other normal GI contents
8. Presystemic metabolism by:
a. Luminal enzymes
b. Gut wall enzymes
c. Bacterial enzymes
d. Hepatic enzymes
Patient Related Factors:
Age
• In infants the gastric pH is high and intestinal surface blood
flow to the GIT is low resulting in alteration of absorption
pattern compared to adults.
• In elderly persons, causes of impaired drug absorption
include altered gastric emptying, decreased intestinal
surface area and GI blood flow, higher incidents of
achlorhydria and bacterial over growth in small intestine.
Gastric Emptying
• The passage of the drug from the stomach to intestine is
called as gastric emptying.
• Gastric emptying is a first order process and can be
described by using three parameters:
• Gastric emptying rate: This is the speed at which the
stomach contents empty into the intestine.
• Gastric emptying time: It is the time required for the gastric
contents to empty into the small intestine.
• Gastric emptying half-life: it is the time taken for half the
stomach contents to empty into the small intestine.
• Gastric emptying can be promoted by taking the drug in
empty stomach while it can be delayed by co-administration
of food.
Rapid gastric empting is advisable when
1. Rapid onset of action is desired E.g. Sedatives
2. Dissolution of drug occurs in intestine E.g. Enteric coated
dosage forms
3. Drugs are unstable in gastric fluids E.g. Penicillin G
4. Drugs irritate gastric mucosa E.g. Aspirin
5. Drug is best absorbed from distal part of small intestine
E.g. Vitamin B12.
Delayed gastric emptying is recommended when:
• The food promotes dissolution and absorption e.g.
Griseofulvin
• Disintegration or dissolution is promoted by gastric fluids
• Drugs are absorbed from the proximal part of the small
intestine
• Drug has absorption window in stomach
Factors affecting GI emptying:
• Volume of meal: Larger the bulk of the meal, longer the
gastric time
• Composition of meal: The rate of gastric emptying for
various food materials is in the following order
carbohydrates > proteins > fats.
• Physical state of meal: Liquid meal takes less time as
compared to solid meals.
• Viscosity of meal: Viscous materials slow GE rate than less
viscous materials.
• GI pH: Gastric emptying is retarded at low stomach pH and
promoted at higher pH.
• Exercise: Vigorous physical training retards gastric
empting.
• Body posture: Gastric emptying is favored while standing
and by lying on right side.
• Emotional state: Stress and anxiety promotes GI motility,
where as depression retards it.
• Drugs: That retards gastric emptying are antacids, anti
cholinergic, narcotic analgesics and tricyclic
antidepressants. And metoclopramide, domperidone and
cisapride stimulate gastric emptying.
• Disease states: diseases retarding GE are: gastroentreritis,
gastric ulcer, diabetes and hypothyroidism. Diseases
promoting GE are: gastrectomy, duodenal ulcer and
hyperthyroidism.
Intestinal Transit
• Since, small intestine is the major site of absorption of most
of drugs, long intestinal transit time is desirable for
complete absorption of drugs.
• The residence time depends upon the intestinal motility that
occurs due to peristaltic contractions.
• Food, decreased digestive secretions, and pregnancy retard
intestinal transit whereas diarrhoea promotes it.
• Delayed intestinal transit is desirable for
– Drugs that dissolve or release slowly from their dosage form
(sustained release products)
– Drugs that dissolve only in intestine (enteric coated formulations)
– Drugs absorbed from specific sites in the intestine (several B
vitamins)
Gastrointestinal pH
• GI pH may influence disintegration, dissolution, absorption
and stability of drug.
• The enteric coated formulation disintegrate only in the
intestine, weak acids dissolve in intestine, weak bases
dissolve in stomach.
• The acidic stomach pH degrades penicillin G and
erthyromycin. Hence they are converted to their stable
derivatives carindacillin and erythromycin estolate
respectively.
Disease states:
GI Diseases
• The altered GI motility affects the absorption.
• Achlorhydria decreases absorption of acidic drugs like aspirin.
• In celiac disease, increased GE rate and GI permeability and impaired
secretion of bile salts is observed which affects the absorption of
lipophilic drugs and vitamins.
• In crohn’s disease, gut wall microbial flora is altered with decrease in gut
surface area and intestinal transit rate.
• GI infections like gastroenteritis and food poisoning result in mal-
absorption.
• Colonic diseases like colitis, amoebiasis and constipation can alter drug
absorption.
• Gastrectomy can result in diarrhea and decreased intestinal transit time.
Cardiovascular Diseases
• Several changes such as edema of intestine, decreased blood
flow to GIT, decreased GE rate, altered GI pH, secretions
and microbial flora influence bioavailability of drugs.
Hepatic Diseases
• Liver cirrhosis influences the bioavailability of drugs that
undergo considerable first pass hepatic metabolism.
Blood flow to the GIT
• GIT is extensively supplied with blood capillaries and
lymphatic system. GI perfusion (blood flow) rate could be a
rate limiting step in the absorption of drugs that have high
permeation rates. Blood flow is also important for actively
absorbed drugs as oxygen and energy is required for their
transportation.
GIT contents
Food drug interactions
• Presence of food may reduce, delay, increase or may not
affect drug absorption. Drugs are better absorbed under
fasting conditions and presence of food retard or prevent
their absorption.
• Food does not influence the absorption of a drug taken half
an hour or more before meal and 2 hour or more after meal.
• Decreased absorption by food may be due to
– Delayed GE or prevents the transit of enteric coated tablets in to
the intestine
– Formation of poorly soluble unabsorbable complex
– Increased viscosity that prevents drug dissolution or diffusion
towards the absorption site.
• Increased absorption by food may be due to
– Increased time for dissolution of poorly soluble drug
– Enhanced drug solubility due to GI secretion like bile
– Prolonged residence time and absorption site contact of the drug
– Increased lymphatic absorption
Food type and Fluid volume
• Components of meal also have an influence on drug
absorption. Fat rich meal aids in solubilization of poorly
soluble drugs. Protein rich meal inhibits absorption of
levodopa a while promotes propranolol bioavailability.
• If drug is taken with large fluid volume better dissolution,
rapid GE and increased absorption is observed.
Interaction of drug with Normal GI constituents
• Mucin interacts with streptomycin and certain quaternary
ammonium compounds to decrease their absorption. It also
acts as barrier to drug diffusion. Bile salts aid solubilization
and absorption of lipid soluble drugs. They decrease
absorption of neomycin and kanamycin by forming water
insoluble complexes.
Drug-Drug Interactions in the GIT
• These interactions can either be physicochemical (involve
adsorption, complexation and pH change) or physiological
(decreased GI transist, increased GE, altered GI metabolism).
Presystemic metabolism (First Pass effects)
• Two main reasons for its decreased bio-availability from
orally administered drugs are: Decreased absorption or First-
pass/pre-systemic metabolism.
• The loss of drug through biotransformation by eliminating
organs during its passage to systemic circulation is called as
first pass or presystemic metabolism.
• Following are primary systems affecting presystemic
metabolism: Lumenal enzymes, Gut wall enzymes, Bacterial
enzymes, Hepatic enzymes.
• Lumenal enzymes: These are enzymes present in gut fluids
and include enzymes from intestinal and pancreatic
secretions. E.g. hydrolases and peptidases
• Gut wall enzymes: Also called mucosal enzymes they are
present in stomach, intestine and colon. E.g. Alcohol
dehydrogenase.
• Bacterial enzymes: GI microflora present in stomach and
small intestine and is rich in colon.
• Hepatic enzyme: several drug undergo first-pass hepatic
metabolism by liver enzymes. E.g. are isoprenaline,
nitroglycerin, morphine, nifedipine, propranolol etc.
Absorption of drug from Non-per oral route
• Buccal/Sublingual Administration
• Rectal Administration
• Topical Administration
• Inhalation Administration
• Intramuscular Administration
• Subcutaneous Administration
• Intranasal Administration
• Intraocular Administration
• Vaginal Administration
(Refer Book: DFD-I, by Dr. D.M.Patel, Nirav Prakashan, Page
no. 166-169)

Das könnte Ihnen auch gefallen