UNIT 2 ELIMINATION and BIOAVAILABILITY & BIOEQUIVALENCE PDF

Download as pdf or txt
Download as pdf or txt
You are on page 1of 207

BIOPHARMACEUTICS &

PHARMACOKINETICS
(BP 604T)
Unit 2(a)
ELIMINATION
SYLLABUS
• Elimination:
– Drug metabolism and basic understanding
metabolic pathways
– renal excretion of drugs,
– factors affecting renal excretion of drugs,
– renal clearance,
– Non renal routes of drug excretion of drugs
METABOLISM
Introduction
• Elimination: removal of a drug from the body & termination
of its action
• Occurs by 2 process- Biotransformation & Excretion

• Biotransformation: Chemical alteration of the drug in body


that converts nonpolar or lipid soluble compounds to polar or
lipid insoluble compounds

• Consequences of biotransformation
• Active drug  Inactive metabolite : Pentobarbitone,
Morphine, Chloramphenicol
• Active drug  Active metabolite: Phenacetin
• Inactive drug  active metabolite: Levodopa
4
Prodrugs
• Inactive drug is converted to active metabolite
• Coined by Albert in 1958
• Advantages:
• Increased absorption
• Elimination of an unpleasant taste
• Decreased toxicity
• Decreased metabolic inactivation
• Increased chemical stability
• Prolonged or shortened action
DRUG METABOLIZING ORGANS
• Liver is the primary site of metabolism of almost all
drugs.
• Liver is relatively rich in possessing a large variety
of enzymes in large amounts.
• Metabolism by organs other than liver is called
extrahepatic metabolism.
• Order of decreasing metabolism:
– Liver>lungs>kidneys>intestine>placenta>adrenals>skin
– Brain, muscles, splee, etc., also metabolize to small
extent.
DRUG METABOLISING ENZYMES
• Metabolising enzymes are versatile and non
specific in metabolising a number of drugs.
• Most metabolising activity in smooth
endoplasmic reticulum and cytosol.
• Enzymes are of two types
– Microsomal (endoplasmic reticulum)
– Non microsomal (cytosol and mitochondria)
PHASES OF METABOLISM
• Phase I
• Functionalization reactions; asynthetic reactions

• Converts the parent drug to a more polar


metabolite by introducing or unmasking a
functional group (-OH, -NH2, -SH).
• Phase II
• Conjugation reactions

• formation of a covalent linkage between a functional


group on the parent compound (or on a phase I
metabolite) with endogenously derived glucuronic
acid, sulphate, glutathione, amino acids or acetate.
PHASES OF METABOLISM
Hydrolytic Reactions
 Esters, amides, epoxides and Oxidation
arene oxides by epoxide hydrase  Aromatic moieties, Olefins
 Benzylic & allylic C atoms
and a-C of C=O and C=N
Phase II -  At aliphatic and alicyclic C
Phase I -
Conjugation  C-Heteroatom system
Functionalization  C-N (N-dealkylation, N-oxide
formation, N-hydroxylation)
 C-O (O-dealkylation)
 S-dealkylation
Drug  S-oxidation, desulfuration
Metabolism  Oxidation of alcohols and
aldehydes, Miscellaneous
 Conjugation
 Glucuronic acid Reduction
 Sulfate, Glycine and otherAA  Aldehydes and ketones
 Glutathione or mercapturic acid  Nitro and azo
 Acetylation, Methylation  Miscellaneous
EXCRETION
Excretion
• Drug excretion is defined as the removal of drugs
from the body .
• Excretion is defined as the process where by
drugs or metabolites are irreversibly transferred
from internal to external environment through
renal or non renal route.
• Excretion of unchanged or intact drug is needed
in termination of its pharmacological action.
• Drugs and their metabolites can leave the body in
urine, bile, sweat, saliva, breast milk, and expired
air. The most important organ for drug excretion
is the kidney.
Drugs are eliminated from the body primarily by
the kidneys.

The principal renal mechanisms that are involved in the


excretion of drugs are:

1. Glomerular filtration
2. Active tubular secretion
3. Active tubular reabsorption
 The ultrastructure of the glomerular capillary wall is
such that it permits a high degree of fluid filtration
while restricting the passage of compounds having
relatively large molecular weights.

 This selective filtration is important in that it


prevents the filtration of plasma proteins (e.g.,
albumin) that are important for maintaining an
osmotic gradient in the vasculature and thus plasma
volume.
 Severalfactors, including molecular size, charge, and
shape, influence the glomerular filtration of large
molecules.

 As the ultrafiltrate is formed, any drug that is free in


the plasma water, that is, not bound to plasma
proteins or the formed elements in the blood (e.g., red
blood cells), will be filtered as a result of the driving
force provided by cardiac pumping.
 Allunbound drugs will be filtered as long as their
molecular size, charge, and shape are not excessively
large.

 Compounds with 20 Å to 42Å may undergo


glomerular filtration.
 The tubular secretion which is carried out at the level
of the proximal tubule is an active process.
 It is carrier mediated process which requires energy
for transportation of compounds against conc.
gradient
Two secretion mechanisms are identified.
System for secretion of organic acids/anions
e.g. Penicillin, salicylates etc
System for organic base / cations
e.g. morphine, mecamylamine hexamethonium
Organic Anion Transport Organic Cation Transport

Acetazolamide Acetylcholine

Bile salts Atropine

Hydrochlorothiazide Cimetidine

Furosemide Dopamine

Indomethacin Epinephrine

Penicillin G Morphine

Prostaglandins Neostigmine

Salicylate Quinine
 In the distal tubule there is passive excretion and re-
absorption of drugs. Drugs which are present in the
glomerular filtrate can be reabsorbed in the tubules. A
reason for this is that much of the water, in the
filtrate, has been reabsorbed and therefore the
concentration gradient is now in the direction of re-
absorption hence drug may be readily reabsorbed.
 Many drugs are either weak bases or acids and
therefore the pH of the filtrate can greatly influence
the extent of tubular re-absorption for many drugs.
When urine is acidic, weak acid drugs tend to be
reabsorbed. Alternatively when urine is more
alkaline, weak bases are more extensively
reabsorbed. Making the urine more acidic can cause
less reabsorption of weak bases or enhanced
excretion.
 In the case of a drug overdose it is possible to
increase the excretion of some drugs by suitable
adjustment of urine pH. For example, in the case of
pentobarbital (a weak acid) overdose it may be
possible to increase drug excretion by making the
urine more alkaline with sodium bicarbonate
injection.
Classification of excretion mechanism
• Types of excretion
• RENAL EXCRETION
• NON RENAL EXCRETION
FACTORS AFFECTING RENAL
EXCRETION OR RENAL CLEARANCE
1. Physiochemical properties of drug
2. Plasma concentration of drug
3. Distribution and binding characteristics of drug
4. Urine pH
5. Blood flow to the kidney
6. Biological factors
7. Drug interactions
8. Disease state
FACTORS AFFECTING RENAL
EXCRETION
• a) Physicochemical Properties Of Drug:
– Lipid solubility: urinary excretion is inversely
related to lipophilicity .
– Stereochemical nature: Chloroquine ,
disopyramide & terbutaline are stereoselectively
secreted by kidneys
PHYSIOCHEMICAL PROPERTIES OF DRUG
 Compounds of weights < 300 Daltons, if watersoluble,
easily excreted by the kidneys.
Drugs or metabolites > 500 Daltons, mainly inbile.

 Drug pKa govern the degree of ionization at theparticular


pH. A polar & ionized drugs poorly reabsorbed
passively excreted rapidly.

 Ionized but lipophilic drug passively reabsorbed.


Unionized but polar excreted in urine.
 Stereoselective protein binding drugs drug
enantiomers exhibit differential filtrationrate.

 Chloroquine, disopyramide and terbutaline


stereoselectively secreted by kidneys. ( active tubular
secretion)

 Stereoselective active tubularreabsorption


glucose and aminoacids.

 Passive reabsorption is unaffected.


FACTORS AFFECTING RENAL
EXCRETION
• b) Plasma Conc. Of Drug: Glomerular filtration
and Reabsorption are directly affected by plasma
concentration.
• c) Distribution and binding characteristics of the
drug :
– Clearance is inversely related to apparent volume of
distribution of drugs.
– A drug with large V d is poorly excreted in urine.
– Drugs restricted to blood compartment have higher
excretion rates
PLASMA CONCENTRATION OF DRUG
 Affects glomerular filtration and reabsorption.(passive)
 Actively reabsorbed drugs excrete in urine only when
their concentration in the glomerular filtrate > active
reabsorption capacity, e.g. glucose.
DISTRIBUTION AND BINDING
CHARACTERISTICS OF DRUG
 Drug with larger Vd poorly excreted in urine.
 Drugs bound to plasma protein cannot be filtered
through the glomerulus.

 ClR = Urine drug concentration × Urine


flowrate Plasma drug concentration
 F u = Cu / C
 ClR = F u × Urine flow rate
 Drugs extensively bound to protein have longerhalf lives
as renal clearance issmall.

 Rena clearance
oxytetracycline (66% unbound) = 99 ml/min.
deoxycycline (7 % unbound) = 16 ml/min.

 Actively secreted drugs are much less affected by protein


binding. E.g. penicillins.
FACTORS AFFECTING RENAL
EXCRETION
• d) Urine pH
– The pH of urine varies between 4.5 – 7.5, thus creating large pH
gradient between urine and plasma. The pH of urine is
dependent upon diet, drug intake and patho -physiology of
patient.
– The relative amount of ionised and unionised drug in the urine
at a particular pH an the % of drug ionised at this pH can be
computed from the Henderson- Hasselbach equations .
– pH = pKa + log ( Ionised drug/ Unionised drug)…Weak acids
– pH = pKa + log ( Unionised drug/ Ionised drug)…Weak bases
– Physicochemical Properties Of Drug:
– Lipid solubility: urinary excretion is inversely related to
lipophilicity .
– Stereochemical nature: Chloroquine , disopyramide &
terbutaline are stereoselectively secreted by kidneys
FACTORS AFFECTING RENAL
EXCRETION
• e) Renal blood flow
– Renal blood flow is important for drugs excreted
by Glomerular filtration only and those are
actively secreted.
• f) Biological factors:
– Renal excretion is 10% lower in females
– Newborns : 30-40% less (attains maturity between
2.5-5 months of age)
– Old age : GFR decreased, excretion decreased,
increased t 1/2 .
FACTORS AFFECTING RENAL
EXCRETION
• g) Drug interactions:
– Alteration of Protein-Drug binding: The renal
clearance of drugs extensively bound to plasma
proteins is increased after displacement with
another drugs.
– E.g . Gentamicin induced nephrotoxicity by
Furosemide . ( Furosemide displaces gentamicin
from protein)
FACTORS AFFECTING RENAL
EXCRETION
• h) Disease states:
– Renal dysfunction : Greatly impairs elimination of
drugs primarily excreted by kidneys
– Disease states :Ureamia
– Impaired Glomerular Filtration
– Accumulation Of Drugs Toxicity .
CONCEPT CLEARANCE
• The clearance concept was first introduced to describe renal
excretion of endogenous compounds in order to measure the
kidney function.
• The term is now applied to all organs involved in drug
elimination such as liver, lungs, the biliary system, etc. and
referred to as hepatic clearance, pulmonary clearance, biliary
clearance and so on.
• The sum of individual clearances by all eliminating organs is
called as total body clearance or total systemic clearance.
• It is sometimes expressed as a sum of renal clearance and
nonrenal clearance.
DEFINITION OF CLEARANCE
• Clearance is defined as the hypothetical volume of
body fluids containing drug from which the drug is
removed or cleared completely in a specific period of
time.
• It is expressed in ml/min and is a constant for any
given plasma drug concentration.

Elimination
Clearance (Cl) = ---------------------------------------
Plasma drug concentration
Renal Clearance
 Renal clearance (ClR) can be defined as “the volume
of blood or plasma which is completely cleared of the
unchanged drug by the kidney perunit time”.
 ClR = Rate of urinary excretion
 Plasma drug concentration

 ClR = Rate of (filtration + secretion – reabsorption)


Plasma drug concentration

 Renal clearance ratio = ClR of drug


ClR of creatinine
MECHANISM OF RENAL CLEARANCE
Renal Renal Mechanism of Renal Example(s)
Clearance Clearance Clearance
(ml/min) Ratio

0 (least value) 0 Drug filtered and Glucose


Reabsorbed completely
< 130 Above 0, Drug filtered and Lipophilic drugs
Below 1 Reabsorbed partially
130 (GFR) 1 Drug is filtered only Creatinine, Inulin
> 130 >1 Drug filtered as well as Polar, ionic drugs
secreted actively
650 5 Clearance equal to renal Iodopyracet
(Highest value) plasma flow rate
DETERMINATION OF CLEARANCE
• One method of quantitatively describing the renal
excretion of drugs is by means of the renal clearance
value for the drug.
• Renal clearance can be used to investigate the mechanism
of drug excretion:
A- If the drug is filtered but not secreted or reabsorbed the
renal clearance will be about 120 ml/min in normal
subjects.
B- If the renal clearance is less than 120 ml/min then we can
assume that at least two processes are in operation,
glomerular filtration and tubular re-absorption.
C- If the renal clearance is greater than 120 ml/min then
tubular secretion must be contributing to the elimination
process.
NON-RENAL ROUTES
• Drugs and their metabolites may also be excreted
by routes other than the renal routes called extra
renal or non renal routes of drug excretion.
• The various such excretion processes are
– Biliary excretion.
– Pulmonary excretion.
– Salivary excretion.
– Mammary excretion.
– Skin excretion.
– Gastrointestinal excretion.
BILIARY SECRETION
• Bile juice is secreted by hepatic cells of the liver.
• In human, the bile flow rate is a steady 0.5 to 1
ml/min .
• Molecular weight between 300-500 Dalton are
excreted both in urine and in bile route.
• Drugs excreted in bile include-
Cromoglycate.Morphine,Indomethachin,Chloram
phenical .
• The phenomenon of drug cycling between
intestine and liver is called as enterohepatic
circulation of drugs .
BILIARY SECRETION

• Compounds excreted by this route are


sodium, potassium, glucose, bilirubin ,
Glucuronide , sucrose, Inulin , muco -proteins
e.t.c .
• Greater the polarity better the excretion. The
metabolites are more excreted in bile than
parent drugs due to increased polarity.
PULMONARY EXCRETION
• Lung is a major organ of excretion for gaseous and
volatile substance.
• Most anaesthetics – extensively eliminated in expired
air.
• Gaseous and volatile substance are absorbed by the
lungs with simple diffusion.
• The excretion of drugs occurs by diffusion of expired
air.
• Drugs having the property of high solubility in blood
and tissue are excreted slowly by lungs
• Examples. - general anaesthetics (halothane) -
salbutamol .
SALIVARY EXCRETION
• Excretion of drugs in saliva is a passive diffusion
process.
• The pH of saliva varies from 5.8 to 8.4 .
• Unionized lipid soluble drugs are excreted passively.
• The bitter after taste in the mouth of a patient is
indication of drug excreted.
• Some basic drugs inhibit saliva secretion and are
responsible for mouth dryness .
• Basic drugs secreted more than acidic drugs E.g.
Lithium, Tetracycline Metronidazole Pyrizinamide
Penicillin Ethosuximide Phenytoin Digoxin
MAMMARY EXCRETION
• Excretion of drug is important in breast-feeding infants .
• pH of milk varies 6.4 to 7.6.
• Mammary excretion Weakly basic drugs concentrate more
in milk. E.g. Chloramphenicol , tetracyclines ,
ergotamine,morphine etc.
• Drug Effect- Tetracycline Permanent staining on teeth
• Sulphonamides - Kernicterus
• Penicillin- Allergy
• Ampicillin -Diarrhea
• Dapsone -Hemolytic anemia
• Phenobarbitone -Drowsiness
• Theophylline -Restlessness
SKIN EXCRETION
• Drugs excreted through skin via sweat.
• Drugs and their metabolites excreted through
mechanisms of passive diffusion.
• Hypersensitivity reactions,urticaria and
dermatitis occurs.(Bromides, Salicylic acid,
Arsenic Salts ).
• Silver salts may discolor the skin.
• Compounds like benzoic acid, salicylic acid,
alcohol and heavy metals like lead, mercury and
arsenic are excreted in sweat.
GASTROINTESTINAL EXCRETION
• Excretion of drugs through GIT usually occurs
after parenteral administration .
• Water soluble and ionized from of weakly acidic
and basic drugs are excreted in GIT.
• Drugs excreted in GIT are reabsorbed into
systemic circulation & undergo recycling
• E.g . Nicotine ,quinine, Streptomycin ,Neomycin ,
Bacitracin , Cholestyramine , Erythropoitein ,
Chlorpromazine ,Corticosteroids are excreted in
stomach .
EXCRETION PATHWAYS, TRANSPORT
MECHANISMS & DRUG EXCRETED.

Excretory Mechanism Drug Excreted


route
Urine GF, ATS, PTR Free, hydrophilic, unchanged drugs/
metabolites of MW< 300
Bile Active secretion Hydrophilic, unchanged drugs/
metabolites/ conjugates of MW
>500
Lung Passive diffusion Gaseous &volatile, blood &
tissue insoluble drugs
saliva Passive diffusion Free, unionized, lipophilic drugs. Some
Active transport polar drugs
Milk Passive diffusion Free, unionized, lipophilic drugs (basic)
Sweat Passive diffusion Free, unionized lipophilic drugs
BIOPHARMACEUTICS &
PHARMACOKINETICS
(BP 604T)
Unit 2(b)
BIOAVAILABILTY AND
BIOEQUIVALENCE
SYLLABUS
• Bioavailability and Bioequivalence:
– Definition and Objectives of bioavailability
– absolute and relative bioavailability
– measurement of bioavailability
– in-vitro drug dissolution models
– in-vitro-in-vivo correlations
– bioequivalence studies
– methods to enhance the dissolution rates and
bioavailability of poorly soluble drugs
Introduction
• The therapeutic effectiveness of a drug depends

 ability of the dosage form to deliver the medicament to


its site of action
 at a rate & amount sufficient to elicit the desired
P’cological response.

• This attribute of the dosage form is referred to as


physiological availability or bioavailability.

3/29/2020 3
Introduction

• Bioavailability : Rate and extent of absorption of


unchanged drug from its dosage form.

• A rapid onset of action is desired - in the treatment of


acute conditions like asthma, pain, etc.

• A slower absorption – to prolong the duration of


action and to avoid first pass effects.

• Factors affecting BA- Pharmaceutical factors;


• Patient related factors;
• Route of administration

3/29/2020 4
Introduction

• The influence of route of administration on drug’s


bioavailability is generally in the foll. order

Parenteral > oral > rectal > topical

• Most drugs are administered orally, for reason of


stability and convenience.

• The dose available to patient – Bioavailable dose.

• The amount of drug that reaches the systemic


circulation – systemic availability
3/29/2020 5
Introduction
• Bioavailable fraction (F), refers to the fraction of
administered dose that enters the systemic circulation.

F = Bioavailable dose
Administered dose

3/29/2020 6
Objectives of bioavailability studies

Important in

1. Primary stages of development of a suitable dosage


form for new drug entity.

2. Determination of influence of excipients, patient


related factors and interaction with other drugs on the
efficiency of absorption.

3/29/2020 7
Objectives of bioavailability studies

3. Development of new formulations of the existing


drugs.

4. Control of quality of a drug product during early stages


of marketing (to determine the influence of processing
factors, storage & stability on absorption).

5. Comparison of availability of drug substance from


different dosage form or from the same dosage form
produced by different manufacturers.
3/29/2020 8
Considerations in bioavailability studies

• Bioavailability – absolute vs. relative

• Single dose vs. multiple dose

• Human volunteer – healthy subject vs. patients

3/29/2020 9
Bioavailability – Absolute vs. Relative
• Absolute bioavailability : When the systemic
availability of the drug administered orally is
determined in comparison to its i.v. administration.

3/29/2020 10
• Relative bioavailability : when the systemic
availability of the drug administered orally is
compared with that of an oral standard of the
same drug (Aq or non aq Sol or Suspension).
• Symbol Fr

3/29/2020
3/29/2020 12
Single dose vs. multiple dose studies

Single dose Multiple dose


• Very common & easy • Difficult to control

• Less exposure to drug & • More exposure to drug


less tedious & tedious

• Difficult to predict • Time consuming


steady state
characteristics &
intersubject variability

3/29/2020 13
Single dose vs. multiple dose
• Multiple dose study has several advantages like

1. More accurately reflects the manner in which the


drug should be used (clinically).

2. Drugs levels are higher due to cumulative effect


which makes its determination possible even by
less sensitive analytical methods.

3/29/2020 14
Single dose vs. multiple dose

3. Better evaluation of the performance of controlled


release formulation is possible.

4. Small inter subject variability is observed which


allows use of fewer subjects.

5. Nonlinearity in pharmacokinetics, if present, can be


easily detected.

3/29/2020 15
Human volunteer – healthy subject vs. patients

• Ideally, the bioavailability study should be carried


out in patients for whom the drug is intended to be
used.

• Advantages
1. Patient is benefited from the study.

2. Reflects better therapeutic efficacy of drug.

3/29/2020 16
Human volunteer – healthy subject vs. patients

3. Drug absorption pattern in disease state can be


evaluated.

4. Avoids the ethical quandary of administering drug


to healthy subjects.

3/29/2020 17
Human volunteer – healthy subject vs. patients

• There are some drawbacks of using patients as


volunteers.
• Stringent conditions such as fasting state required is
difficult to be followed by the patients.
• Disease, other drugs, physiological changes – modify
drug absorption pattern

 Studies are therefore performed in young (20-40 yrs.),


healthy males adult volunteers (body weight within
narrow range).

3/29/2020 18
Human volunteer – healthy subject vs. patients

• Female volunteers are used only when drugs such as


oral contraceptives are to be tested.

• No. of subject to be selected- extent of inter subject


variability –minimum required to obtain reliable data.

• They must be informed about the importance of


 Study
 conditions to be followed
 Possible hazards if any.
3/29/2020 19
Human volunteer – healthy subject vs. patients

• Consent of volunteers

• Medical examination should be performed to exclude


subject of any abnormality
• Volunteers instructed to abstain from any medication
for atleast a week
• Fast overnight prior to & for a minimum of 4 hrs after
dosing

• Drug washout period for min. of ten biological half lives


must be allowed for between two studies in same
subject.
3/29/2020 20
Measurement of bioavailability

• Pharmacokinetic methods ( indirect ) –p’cokinetic


profile reflects therapeutic effectiveness of drug
1. Plasma level time studies
2. Urinary excretion data

• Pharmacodynamic methods ( direct ) –measurement


of drug effect on physiological process as a
function of time
1. Acute pharmacological response
2. Therapeutic response

3/29/2020
Plasma level time studies
• Plasma level time studies or The plasma concentration – time curve
or blood level curve.

• A direct relationship exists concentration of drug at the site of


action & concentration of drug in the plasma.

• Assumption: two dosage forms that exhibit superimposable plasma


level time profiles in a group of subjects should result in identical
therapeutic activity

• Serial blood samples for a period of 2-3 biological half life are
taken after drug administration

• analyzed for drug concentration.

3/29/2020 22
• Plot of drug concentration vs corresponding time
of sample collection

• I.V dose- sampling start within 5 min of


administration

• Subsequent sampling: at 15 min intervals

• At least 3 points should be taken (1 compartment


model)
• 5-6 Sample points (2 compartment model)
3/29/2020 23
3/29/2020
3/29/2020 25
3/29/2020 26
3/29/2020 27
3/29/2020 28
3/29/2020 29
3/29/2020 30
3/29/2020 31
3/29/2020 32
3/29/2020 33
3/29/2020 34
3/29/2020 35
3/29/2020 36
3/29/2020 37
Parameters determined
• AUC or Extent of absorption can be measured by 3 method
1.Planimeter
Instrument for mechanically measuring the area

2. Cut & weigh method


AUC is cut & weighed on analytical balance. The weight obtained is
converted to proper unit by dividing it by the wt of a unit area of
same paper.

3. Trapezoidal method

3/29/2020 38
3/29/2020 39
3/29/2020 40
USP Def.
According USP IVIVC is
“the establishment of a rational relationship
between a biological property or a parameter
derived from a biological property produced by a
dosage form, and a physicochemical property or
characteristic of the same dosage form”.

biological property physicochemical property

3/29/2020 41
parameter derived from a biological property

parameter derived from physicochemical


characterstics

• parameter derived from the biological property


is AUC or Cmax,
• physicochemical property is the in vitro
dissolution profile

3/29/2020 42
Objective of IVIVC
• To enable the dissolution test to serve as a surrogate for
in vivo bioavailability studies in human beings
• To ensure batch to batch consistency in physiological
performance of drug pdt by use of in vitro values
• To serve as tool in development of new dosage form
• To assist in validating dissolution specifications
• IVIVC allows dosage form optimization with the fewest
possible trials in man

3/29/2020 43
In vitro drug dissolution rate &
bioavailability
• Best way of assessing therapeutic efficacy of drug- in
vivo determination of bioavailability (intro of new
formulation in market)
• Monitoring batch to batch consistency through use of in
vivo test- costly, tedious & time consuming
• In vitro dissolution test- substitute to in vivo
bioavailability
• To quantitatively assure about the biological availability
of a drug from its formulation

3/29/2020 44
In vitro dissolution testing models
• In vitro test- useful if it predict in vivo behaviour to such an
extent that no need to perform in vivo bioavailability test.
• Factors to consider while designing dissolution test

1. Related to Apparatus- Design, size of container, Shape, Nature


of agitation, speed of agitation

2. Related to dissolution fluid- Composition, Viscosity, Volume,


Temperature, Sink/ Non sink condition

3. Process parameters- sampling techniques, changing dissolution


fluid
3/29/2020 45
3/29/2020 46
3/29/2020 47
3/29/2020 48
3/29/2020 49
3/29/2020 50
Apparatus 4: Flow
Through Cells

51
3/29/2020 52
• Close/open mode
• advantage of unlimited medium supply, which is
of particular interest for the dissolution of
poorly soluble drugs
• Tablets, sugar-coated tablets, suppositories,
soft-gelatin capsules, semisolids, powders,
granules, and implants
• medium flows through the cell from bottom to
top of the cell
• Volumes used: 500 to 4000 mL
• Feasibility for automation of apparatus
3/29/2020 53
3/29/2020 54
3/29/2020 55
3/29/2020 56
3/29/2020 57
3/29/2020 58
3/29/2020 59
3/29/2020 60
3/29/2020 61
Parameters used for correlating in vitro
dissolution with plasma data
In vitro dissolution parameters In vivo plasma data parameters

Time for specific amount of drug to AUC, C max


dissolve

Amount dissolved at a specific time point Fraction absorbed, Ka

Mean dissolution time Mean Residence time, Mean absorption


time

Parameters estimated after modelling Concentration at time t; Amount absorbed


dissolution process at time t

3/29/2020 62
• Correlation based on Urinary excretion data
Dissolution parameters
correlated to

Amount of drug excreted unchanged in urine;


Cumulative amount of drug excreted as a function of time

• Correlation based on Pharmacological Response


Dissolution parameters
correlated to

LD 50

3/29/2020 63
Level A Correlation
• Highest category of correlation
• Point to point relationship b/w in vitro
dissolution & in vivo rate of absorption
• in vitro dissolution & in vivo rate of absorption
rate curves are super imposable
• in vitro dissolution curve serve as a surrogate for
in vivo performance.

3/29/2020 64
3/29/2020 65
• Any change in manufacturing procedure/
modification in formula – justified without need
for additional human studies

• In vitro dissolution serves as in vivo indicating


quality control procedure for predicting dosage
form’s performance

3/29/2020 66
Level B Correlation
• Principle of statistical moment analysis
• MDT vitro is compared to either MRT or MDT vivo

• Not a point to point correlation since number of


different in vivo curves will produce similar MRT
values
• A level B correlation does not uniquely reflect
the actual in vivo plasma level curves
• Cant rely to justify changes in manufacturing/
modification in formula
3/29/2020 67
3/29/2020 68
3/29/2020 69
3/29/2020 70
Level C Correlation
• Single point correlation
• Relates one dissolution time point (t 50% ) to one
P’cokinetic parameter such as AUC, tmax or Cmax
• Weakest level of correlation
• A Level C correlation does not reflect the
complete shape of the plasma concentration-
time curve,
• Useful as a guide in early stages of formulation
development (when pilot formulations are being
selected)
3/29/2020 71
3/29/2020 72
3/29/2020 73
Multiple Level C Correlation
• Involve one or several pharmacokinetic
parameters (AUC, Cmax or any other suitable parameter)
to the amount of drug dissolved at various time
points.
• Should be based on at least three dissolution
time points covering the early, middle and later
stages of the dissolution profile.

3/29/2020 74
3/29/2020 75
• BCS is a fundamental guideline for determining
the conditions under which in vitro/in-vivo
correlations are expected.

• also used as a tool for developing in-vitro


dissolution specification

• IVIVC: normally expected for highly permeable


drugs or drugs under dissolution rate-limiting
conditions.

3/29/2020 76
3/29/2020 77
3/29/2020 78
Bioequivalence

3/29/2020 1
3/29/2020 2
3/29/2020 3
• Pharmaceutical Equivalents: Drug products are considered
pharmaceutical equivalents if they contain the same active
ingredient(s), are of the same dosage form, route of
administration and are identical in strength or concentration

• Pharmaceutical Alternatives : Drug products are considered


pharmaceutical alternatives if they contain the same therapeutic
moiety, but are different salts, esters, or complexes of that
moiety, or are different dosage forms or strengths

(e.g., tetracycline hydrochloride, 250mg capsules vs. tetracycline


phosphate complex, 250mg capsules)

3/29/2020 4
3/29/2020 5
3/29/2020 6
3/29/2020 7
3/29/2020 8
3/29/2020 9
3/29/2020 10
3/29/2020 11
3/29/2020 12
3/29/2020 13
3/29/2020 14
3/29/2020 15
BE Study Designs
• Standard BE Study design (non replicate)

2 formulations are compared in a randomized


two period, two sequence, single dose
crossover design

Latin square cross over design

3/29/2020 16
3/29/2020 17
3/29/2020 18
3/29/2020 19
3/29/2020 20
3/29/2020 21
3/29/2020 22
Alternative BE study designs
• Parallel design: used for drug with very long half
life
• Replicate design: for substances with variable
pharmacokinetic characteristics
Partial replicate : 1 treatment (T/R) administered
to same subject on 2 separate occasion
Full replicate: both treatments of T & R are
administered to same subject on 2 separate
occasion
3/29/2020 23
3/29/2020 24
3/29/2020 25
• Registration of a generic formulation requires
demonstration of bioequivalence with the accepted
reference formulation of same drug.

• P’cokinetic parameter: Cmax; AUCo-t ; AUC 0-infinity

• Bioequivalence is established if 90% confidence interval


falls within the range of 80% -125%

3/29/2020 26
• 2 sequential clinical trials are conducted for BE
determination:
Pilot BE study
Small number of subjects
Used to validate analytical methodology, assess
variability,
optimize sample collection time intervals
Provide other info
Pivotal BE study
Demonstrate BE using appropriate number of
subjects determined by pilot study
3/29/2020 27
3/29/2020 28
3/29/2020 29
3/29/2020 30
3/29/2020 31
Sampling
• The sampling scheme should be frequent
enough to define the absorption phase, and the
elimination phase during a time course in the
body

• Withdrawal, storage and handling of sample

3/29/2020 32
2. Subjects:
A. Selection of subjects
 Aim to minimize variability and permit detection of differences
between pharmaceutical products.
 The studies should normally be performed with healthy
volunteers.
 They should be screened for suitability by means of clinical
laboratory tests, review of medical history, and medical
examination, alcohol intake, smoke, drug abuse.
Standardization of the study:
 The test conditions should be standardized in order to minimize
the variability of all factors involved.
 Standardization of the diet, fluid intake and exercise is
recommended.
 The subjects should not take other medicines during a suitable
period before and during the study.
C. Inclusion of patients:
• If the investigated active substance is known to have adverse effects
and the pharmacological effects or risks are considered unacceptable
for healthy volunteers it may be necessary to use patients instead,
under suitable precautions and supervision.
D. Genetic phenotyping:
• Phenotyping of subjects should be considered for exploratory BA
studies as well in parallel studies.
• If a drug is known to be subject to major genetic polymorphism,
studies could be performed in panels of subjects of known
phenotype or genotype.
3. Characteristics to be investigated:
• In most cases evaluation of bioavailability and bioequivalence will be
based upon the measured concentrations of the parent compound.
• In some situations, measurements of an active or inactive metabolite
is carried out.
• The plasma concentration versus time curves are mostly used to
assess extent and rate of absorption.
• The use of urine excretion data may be advantageous in determining
the extent of drug input in case of products predominately excreted
renally.
• Specificity, accuracy and reproducibility of the methods should be
sufficient.
4.Chemical analysis:
• It is conducted according to the applicable principles of Good
Laboratory Practice (GLP).
• Determination of the active moiety and/or its biotransformation
product(s) in plasma, urine or any other suitable matrix must be well
characterized, fully validated and documented to yield reliable results
that can be satisfactorily interpreted.
• The bioanalytical methods used to determine the active principle
and/or its biotransformation products in plasma, serum, blood or
urine or any other suitable matrix must be well characterized,
fully validated and documented

• Performance and reliability of analytical results are


(1) stability of the analyte (s) in the biological matrix under
processing conditions and during the entire period of storage
(2)specificity/ selectivity
(3)precision & accuracy
(4) Sensitivity
(5) recovery
(6)range & linearity
(7) Analytical system availability

3/29/2020 36
5 Reference and test product:
• The choice of reference product should be justified by the applicant
and agreed upon by the regulatory authority.
• The test products used in the biostudy must be prepared in
accordance with GMP-regulations.
6.Data analysis:
• To quantify the difference in bioavailability between the reference
and test products and to demonstrate any clinically important
difference.
7.Statistical analysis:
• The statistical analysis (e.g. ANOVA) should take into account
sources of variation that can be reasonably assumed to have an
effect on the response variable.
• Pharmacokinetic parameters derived from measures of
concentration, e.g. AUC, Cmax should be analyzed using ANOVA.
FORMAT AND CONTENT OF THE REPORT ON
BIOEQUIVALENCE STUDIES
• should give the complete documentation of its
protocol,
• conduct and evaluation complying with Good
Clinical Practice (GCP) and Good Laboratory
Practice (GLP) rules

• The responsible investigator(s) should sign for


their respective section of the report.

3/29/2020 38
• Name and affiliations of the responsible
investigator(s)
• site of the study
• period of execution
• names and batch numbers of the pharmaceutical
products used in the study
• analytical validation report
• Results of in vitro dissolution tests
• procedure for calculating the parameters used
• pharmacokinetic model and computing procedure
• Drop-out and withdrawal of subjects

3/29/2020 39
METHODS TO ENHANCE THE DISSOLUTION RATES AND
BIOAVAILABILITY

You might also like