SrinivasaReddyEdurur
972 views
55 slides
May 23, 2021
Slide 1 of 55
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
About This Presentation
This document explained in detail about Bioequivalence studies
Size: 2.22 MB
Language: en
Added: May 23, 2021
Slides: 55 pages
Slide Content
Bioequivalence Studies
Mr. Srinivasa Reddy. Edururi
The main objective
Purpose of Bioequivalence studies
Phases of Bioequivalence studies
Objective
Introduction
Drug discovery & development
Generic Drugs
Purpose of Bioequivalence studies
Phases of bioequivalence study
Clinical
Bioanalytical
Pharmacokinetics & statistics
Regulatory submission (Module-5)
Contents
The life cycle of any drug involves four phases
discovery and research,
development,
regulatory review and approval, and
commercialization/ marketing.
It takes approximately 10-12 years from the initiation and identification
of the drug target to bring the drug into the market
Drug discovery & development
Drug discovery & development
Discovery & Research
identify a suitable target
understand how the disease occurs at the
molecular, cellular, and genetic levels
understanding how the target plays a role
in the disease process
testing of the target against different
known and new compounds to identify
either one or several ‘lead compounds’
Drug Development
testing and optimization of the selected
compounds to identify the ‘lead structure
or candidate drug’ which might be the
most effective
Testing is done in cells (in vitro) and in
animals (in vivo) to study metabolism
(pharmacodynamics and
pharmacokinetics), safety, toxicity,
dosage, and efficacy.
clinical phase of development, the efficacy
and safety of a drug candidate
is investigated in patients
Regulatory approval
applications with all the necessary
information, including quality, preclinical and
clinical data collected during development of
the product, are submitted to the relevant
regulatory authorities
obtain approval to market the drug in their
jurisdictions (e.g., a New Drug Application
(NDA) in the US, and a Marketing
Authorisation Application (MAA) in the EU).
Marketing
marketing or commercialization of the drug
once it has been approved
The drug manufacturer must submit
marketing authorization applications in every
country or territory in which it wants to sell
the drug.
To continue evaluating the safety and efficacy
of the marketed drug, and for further
development, Phase 4 clinical trials are
conducted by the manufacturer
Clinical Trial Phases
Brand Vs. Generic drugs
Brand Name Drugs
Also called innovator drugs
Initially marketed as new
chemical entities
Supplied by one drug company
(innovator manufacturer)
Sold under companies trade
mark name
Cost is more
Generic Name Drugs
Copies of brand name drugs
Produced after the original
patent expires
May be supplied more than
one company
May be sold under active
ingredient (s) name (s)
Cost is less
Same active ingredient(s)
Same route of administration
Same dosage form
Same strength
Same conditions of use
Compared to brand product
What are the requirements for a
generic drug?
Brand Name Drug Generic Drug
NDA Requirements ANDA Requirements
1. Chemistry 1. Chemistry
2. Manufacturing 2. Manufacturing
3. Controls 3. Controls
4. Labeling 4. Labeling
5. Testing 5. Testing
6. Animal Studies
7. Clinical Studies 6. Bioequivalence
8. Bioavailability
New Drug Vs Generic
Bioavailability
the rate and extent to which the active ingredient or active moiety is
absorbed from a drug product and becomes available at the site of
action.
Bioequivalence
the absence of a significant difference in the rate and extent to which
the active ingredient or active moiety in pharmaceutical equivalents
or pharmaceutical alternatives becomes available at the site of drug
action when administered at the same molar dose under similar
conditions in an appropriately designed study
Bioavailability / Bioequivalence
When do we do bioequivalence studies?
Clinical Service Form to Final Market Form
Change of formulations (capsules to tablet)
Generic Formulations
Change of Process or manufacturing site (some times)
Purpose Bioequivalence studies
Clinical Phase
Protocol preparation to sample transfer to Bioanalytical
Bioanalytical Phase
Method development to data transfer to Pharmacokinetics
Pharmacokinetics & Statistics
Pharmacokinetic and statistical evalusiton
Phases of Bioequivalence study
Protocol Preparation
Ethics committee approval
Informed consent process
Selection of subjects
Test article Packaging
Drug administration
Sample collection/separation/Storage
Adverse Event Monitoring
Transfer of samples to Bioanalytical Laboratory
Clinical Phase
Protocol contents
♦„A document that describes the
objective(s), design, methodology,
statistical consideration and
organisation of a trial. It usually
gives the background and rationale
of the trial …“
Ref.: ICH GCP Guidance
Ethics committee approval
Who: An independent body (a review board or a committee,
institutional, regional, national, or supranational), constituted of
medical professionals and non-medical members.
The composition may be as follows :-
Chairperson
1-2 basic medical scientists (preferably one pharmacologists).
1-2 clinicians from various Institutes
One legal expert or retired judge
One social scientist / representative of non-governmental voluntary agency
One philosopher / ethicist / theologian
One lay person from the community
Member Secretary
Ethics committee approval
Responsibility: to ensure the protection of the rights, safety and well-
being of human subjects involved in a trial.
reviewing and approving / providing favourable opinion on, the trial
protocol,
the suitability of the investigator(s), facilities, and
the methods and material to be used in obtaining and documenting
informed consent of the trial subjects.
Informed consent is a PROCESS
Involves
Providing all relevant information to the volunteer/ patient
The patient/ volunteer understanding the information provided
Voluntarily agreeing to participate
Informed consent is a communication process :
between the researcher and the participant
starts before the research is initiated
continues throughout the duration of the study
Consent must be in a language the subject understands
Personally sign and date by subjects
What is informed consent?
Objective: minimising interindividual variability in order to
detect product differences.
The inclusion and exclusion criteria should be clearly stated
in teh protocol
General screening
The subjects should be screened for suitability by means of clinical
laboratory tests, a medical history, and a physical examination.
ECG, Chest-X ray, etc..
Selection / Enrollment of subjects
Age & BMI
18 years of age or older, between 18.5 and 30 kg/m
2
Gender
Males or Females
Smoking status
Smokers or Non smokers
Number of subjects (sample size calculation)
12, 24, ……
Selection / Enrollment of subjects
Drug administration as per randomization
Generally administered at room temperature
Fluid for drug administration
Water
Apple sauce
Other fluids
Quantity of fluid
Drug administration
USFDA about 8 ounces (240 mL) of water
EMEA Administered with a standardised volume of fluid (at least 150 ml)
CDSCO Not Specified
Canada
The dose should be taken with water of a standard volume (150 – 250
millilitres) and at a standard temperature
Furthermore, the drugs should be administered at approximately the same
time on each study day.
Type of sample
Blood, urine, etc.
Sample collection at specified time points
0.0, 0.5, 1.0, 1.33, …………………..
Centrifugation & separation
Plasma
Serum
Storage of samples
Not to cause significant degradation
-30˚C or -80˚C
Sample collection/separation/Storage
Subject Well-being
Adverse Event documentation
Evaluation of seriousness (Mild, Moderate, severe)
Evaluation of relation to investigative drug
Follow-up of Adverse Events
Concomitant Medications
Serious Adverse Events
Hospitalization (if required)
Adverse Event Monitoring
Sorting of samples (subject wise)
Transfer of samples
Storage conditions shall be maintained (dry ice)
Sample transfer to Bioanalytical
LC-MS/MS or HPLC
Method Development
Method Validation
Routine sample analysis
Bioanalytical Phase
It describes the series of steps and tests performed to determine the
most effective and efficient means of quantitating the specific
compound of interest.
Parameters to be evaluated in MD
Collection of Literature
Reference standards
Selection of CC range
Tuning of molecule in MS
Selection of Internal standard
Selection of Mobile phase
Selection of column
Role of pH
Method Development
Literature search and prior methodology
Multiple reaction monitoring mass spectrometry (MRM)
& Optimization
Development of Sample Preparation Method
Development and optimization of the LC method
SPE Method
Validation of the method
Sample Analysis
LLE Method
Protein Precipitation
Method
Incurred Sample
Repeat Analysis
Routine Batch
Sample Analysis
Repeat
Analysis
Bioanalytical method validation includes all of the procedures that
demonstrate that a particular method used for quantitative measurement of
analytes in a given biological matrix, such as blood, plasma, serum, or urine,
is reliable and reproducible for the intended use.
Full validation:
performed when implementing methods for the first time, or when additional
analytes or metabolites are added for quantitation
•Partial validation:
transfer of the bioanalytical method to another laboratory, change in
equipment, calibration concentration range, limited sample volume, another
matrix or species, change in anticoagulant, sample processing procedure,
storage conditions etc.
•Cross Validation:
Where data are obtained from different methods within and across studies
or when data are obtained within a study from different laboratories,
applying the same method, comparison of those data is needed and a cross
validation of the applied analytical methods should be carried out.
Method Validation
Calibration standards and quality control
samples (QC)
Authenticated analytical reference standard
should be used to prepare (separately)
solution of known concentration
certified reference standards
Never from a marketed drug formulation
commercially supplied reference standards
other material of documented purity
Validation
methods 31
Reference Standard
It is a general method for determining the concentration of a substance in an unknown
sample by comparing the unknown to a set of standard samples of known
concentration.
Instrumental response i.e., the analytical signal, changes with the concentration of the
analyte. A series of standards across a range of concentration near the expected
concentration of analyte in the unknown will give a linear plot.
The calibration curve should be prepared in the same biological matrix (e.g. plasma )
as the sample in the intended study by spiking with known concentration of the
analyte (or by serial dilution).
Calibration Curve
LLOQ: It is the lowest concentration of the standard curve that can be
measured with acceptable accuracy and precision.
ULOQ: The highest standard will define the upper limit of quantification
of an analytical method.
QC samples: These are standards of spiked samples with known
concentrations that provide a method of control (assessing integrity)
for each assay & they must meet the acceptance criteria related to
accuracy and precision.
This can be accomplished by analysis of replicate sets of analyte
samples of known concentration i.e., QC samples from an equivalent
biological matrix.
At a minimum, 3 concentrations representing the entire range of the
standard curve should be studied:
• low QC-within 3 times the LLOQ
• middle QC-approx. 50 to 60% of ULOQ),
• high QC- approx. 70% of ULOQ (near the upper boundary of the
CC curve).
Response: dependent variable
(peak,area ..)
Y (observed)
Y
X
y = ax + b
Independent variable:
exactly known
concentrations
Calibration curve
x1
y1
xn
Yn
Response: dependent variable
Y (observed)
Y
X
y = ax + b
Independent variable:
X
estimated concentration
^
Calibration curve
x1
y1
xn
Yn
x
Response
x
Response
GOOD BAD
^ ^
Calibration curve
Parameters FDA EMEA
Selectivity Biological matrix [plasma (at least one
heamolysed & lipemic), urine, or
other matrix] should be obtained
from at least 6 sources.
At least 6 individual sources.
Response of interfering peak
should be ≤ 20% of the LLOQ for
analyte and ≤ 5% for the IS.
Accuracy The mean value (min five
determinations) should be within 15%
of the actual value except at LLOQ,
where it should not deviate by more
than 20% (minimum Three
Concentrations)
The mean conc. (min five
determinations) should be
within 15% of the nominal values
for the QC samples, except for
the LLOQ which should be within
20% of the nominal value
(minimum four concentrations).
Precision The within & between run CV value
should be ≤ 15% for the QC samples
7 for LLOQ ≤ 20%.
The within & between run CV
value should be ≤ 15% for the
QC samples & for LLOQ ≤ 20%.
Calibration/S
tandard
Curve
It should consist of a blank sample, a
zero sample, & 6 to 8 non-zero
samples covering the expected range,
including LLOQ.
The back calculated conc. of the
CC should be within ±15% of the
nominal value & for LLOQ it
should be within ±20%.
CC/Conc.
Response
•20% deviation of the LLOQ from nominal conc.
•15% deviation of standards other than LLOQ from nominal conc.
Parameters FDA EMEA
LLOQ
The analyte response at the LLOQ
should be at least 5 times the
response compared to blank response
& analyte peak response reproducible
with a precision of 20% and accuracy
of 80-120%.
The analyte signal of the LLOQ
sample should be at least 5
times the signal of a blank
sample.
Freeze and
Thaw
Stability
Analyte stability should be
determined after three freeze and
thaw cycles.
The number of cycles in the
freeze-thaw stability should
equal or exceed that of the
freeze/thaw cycles of study
samples.
Short-Term
Temperature
Stability:
3 aliquots of each of the low and high conc. should be thawed at Room
temp and kept at room temperature from 4 to 24 hours and analyzed.
Long-Term
Stability
The storage time in a long-term
stability evaluation should exceed the
time between the date of first sample
collection and the date of last sample
analysis.
stability at each temperature at
which study samples will be
stored should be determined.
Parameters EMEA
Stock
Solution
Stability
Should be evaluated at RT for at least 6 hr.
If the stock solutions are refrigerated or frozen for the relevant period,
the stability should be documented
Post-
Preparative
Stability
The stability of the drug and the IS in the processed samples should be
assessed over the anticipated run time for the batch size
Whole Blood
Stability
Should demonstrate sample integrity from the time of blood sample is
drawn to the time the plasma/serum is obtained.
Recovery Recovery of the analyte need not be 100%, but the extent of recovery of
an analyte and of the IS should be consistent, precise, and reproducible.
Big Batch Should demonstrate accuracy and precision of QC samples with at least
one run in a size equivalent to prospective study samples analytical run.
Carry-over
Carry over in the blank sample following the high concentration standard
should not be greater than 20% of the LLOQ and 5% for the IS.
Dilution
Integrity
Accuracy and precision should be within the set criteria, i.e. within ±15%.
Dilution integrity should cover the dilution applied to the study samples
Matrix Effect
at least 6 lots of blank matrix(heamolysed & hyperlipidaemic) from
individual donors. The CV should not be greater than 15 %.
Analysis of study samples
An analytical run consists of the
•Blank sample (processed matrix sample without analyte and without IS)
•A zero sample (processed matrix with IS)
•Calibration standards at a minimum of 6 concentration levels
•At least 3 levels of QC samples (low, medium and high) in duplicate (or at
least 5 % of the number of study samples, whichever is higher)
•Study samples to be analyzed.
Possible reasons for reanalysis of study samples and criteria to select the
value to be reported should be predefined in the protocol, study plan or
SOP, before the actual start of the analysis of the samples.
Reanalysis of study samples
Differences for instance in protein binding, back-conversion of known and
unknown metabolites, sample inhomogeneity or concomitant medications,
may affect the accuracy and precision of the analyte in such samples during
processing and storage.
•10% of the samples should be reanalyzed in case the number of samples
is less than 1000 samples.
•5% of the number of samples exceeding 1000 samples.
The concentration obtained for the initial analysis and the concentration
obtained by reanalysis should be within 20% of their mean for at least 67% of
the repeats.
Incurred samples reanalysis
the pharmacokinetic analysis, where pharmacokinetic
parameters derived from the concentrations are
calculated;
Statistics: is the study of the collection, organization,
analysis, interpretation, and presentation of data
Pharmacokinetics & Statistics
Comparison
AUCt The area under the curve (AUC - calculated by the linear trapezoidal rule) from
time zero up to the sampling time for which the last measured concentration
was equal to or larger than LOQ.
AUCinf The AUC from time zero to infinity estimated by adding to AUCt a value equal to
Clast/Kel, where Clast is the measured last quantifiable concentration (occurring
at time Tlast) and Kel is the apparent elimination rate constant (see below).
Cmax The maximum observed concentration of the analyte over the sampling
interval.
Tmax The sampling time at which Cmax occurred.
Kel Apparent first-order terminal elimination rate constant estimated as the
absolute value of the slope of the leastsquares regression line fitted through
the ln(concentration)-time data pairs located on the terminal linear phase of the
ln(concentration)-time profile
Thalf The apparent terminal elimination half-life of the analyte,
calculated as ÷
Pharmacokinetic parameters
The area under the curve (AUC - calculated by the linear trapezoidal rule)
from time zero up to the sampling time for which the last measured
concentration was equal to or larger than LOQ.
Sample time point deviations
AUCt-Area under the curve
Cmax-The maximum observed concentration of the analyte over the sampling interval.
The AUC from time zero to infinity estimated by adding to AUCt a value equal to Clast/Kel,
where Clast is the measured last quantifiable concentration (occurring at time Tlast) and Kel is
the apparent elimination rate constant (see below).
To determine it, we need to calculate slope of the straight line having y-intercept = B. if natural log are used
the rate constant of elimination (b) = negative slope of this straight line.
A key parameter for calculation of AUCinf and Thalf
The AUC from time zero to infinity estimated by adding to AUCt a value equal to Clast/Kel,
where Clast is the measured last quantifiable concentration (occurring at time Tlast) and Kel is
the apparent elimination rate constant (see below).
Thalf-Half-life
The period of time required for the concentration or amount of drug in the body to be reduced
by one-half
Usually consider the half life of a drug in relation to the amount of the drug in plasma.
A drug’s plasma half-life depends on how quickly the drug is eliminated from the plasma.
Formula
Thalf = Ln (2) / Kel
or
Thalf = 0.693 / Kel
Kel = 0.41073
Thalf = 0.693/0.41073
= 1.69
Time Ln Conce
6.00 2.4
7.00 2.2
8.00 2.0
9.00 1.8
10.00 1.7
slope -0.17834
Kel 0.41073
Thalf (T1/2) 1.69
USFDA
The calculated 90% confidence interval for AUC and Cmax should l fall within the
bioequivalence range, usually 80-125%.
EMEA
In studies to determine bioequivalence after a single dose, the parameters to be analysed
are AUC(0-t), or, when relevant, AUC(0-72h), and Cmax.
For these parameters the 90% confidence interval for the ratio of the test and reference
products should be contained within the acceptance interval of 80.00-125.00%.
CDSCO
The calculated 90% confidence interval for AUC and Cmax should l fall within the
bioequivalence range, usually 80-125%.
Canada
The 90% confidence interval of the relative mean AUC
T of the test to reference product should be
within 80.0 % to 125.0 % inclusive.
The relative mean maximum concentration (C
max) of the test to reference product should be between
80.0 % to 125.0 % inclusive.
Acceptable Limits
T/R (%) 80% 125%
Demonstrate BE
Fail to Demonstrate BE
Fail to Demonstrate BIE
Demonstrate BIE Demonstrate BIE
Acceptable Limits