SlideShare a Scribd company logo
1 of 67
Regulatory
Requirements for
Drug Approval
CONTENTS
Introduction
Drug development team
Clinical drug development team
Non-clinical drug development
Various pharmacological approaches to drug discovery
Safety Pharmacology
Toxicological approaches to drug discovery
IND Application
ND Application
Clinical Research protocols
Basic concepts of Biostatistics
Data presentation for FDA submissions
Management of clinical studies
INTRODUCTION
Currently different nations have to follow different
requirements for the regulatory approval of novice (novel)
drug. It is almost difficult for every country to have the same
regulatory approach for the Marketing Authorization
Application (MAA). Therefore it is necessary to have
knowledge about regulatory requirements for MAA of each
country.
Different phases of clinical trials
New Drug Application (NDA) is an application submitted to the
respective regulatory authority for permission to market a new
drug. To obtain this permission a sponsor submits preclinical
and clinical test data for analyzing the drug information and
description of manufacturing procedures. After agency
received the NDA, it undergoes a technical screening. This
process of evaluation is made to ensure that the sufficient
data and the required information have been submitted in
each area justifying the "filling" application form.
Possible action after the review of NDA
Drug Development Team: Drug development is the process
of bringing a new pharmaceutical drug to the market once a
lead compound has been identified through the process of
drug discovery. It includes pre-clinical research on
microorganisms and animals, filing for regulatory status, for an
investigational new drug (IND) to initiate clinical trials on
humans, and may include the steps of obtaining regulatory
approval with a new drug application to market the drug.
The process of drug discovery and development is very long
and needs 10-12 years which includes the close interaction of
large number of scientific disciplines. Most Biotechnology and
Pharmaceutical companies employ teams to mentor the
process of various stages of drug development and making
the drug candidate into therapeutic products
The DDT is comprised of different sub-teams namely drug
discovery team, preclinical drug development team and
clinical drug development team.
(a) Drug Discovery Team: Drug discovery team is a small
group of researchers and is the first team in DDT. The primary
responsibility of this team is to identify lead compounds or
classes of compounds. They screen existing compounds in
the company’s archives or in the libraries of compounds that
are optimized for activity using combinatorial chemistry.
The primary endpoint used in this assessment is the
potency of the various lead compounds in the disease
model. The pharmacology results are shared with the
chemists, who then prepare analogues of the most active
compounds to explore further the structure activity
relationship (SAR). Once a lead compound or class of
lead compounds has been identified, this team commonly
grows to include other scientific disciplines to characterize
more fully the possibility of developing the lead
compounds of interest.
Drug discovery team then includes other experts from various
scientific disciplines such as pharmacology, chemistry,
toxicology, drug metabolism and pharmacokinetics, and bio
pharmaceutics.
(b) Preclinical Drug Development Team:
Once the management accepts a lead compound candidate
as a drug candidate, its further establishment is carried out by
preclinical drug development team (PDDT). This new team
also involves researchers from the various scientific
disciplines. Some companies have Separate groups to support
discovery research and for conducting non-clinical and clinical
developmental studies.
In addition to the drug discovery team members, the PDDT has
a number of new players that include:
1. A management: They assign team leader and coordinator
to the team who responsible for the development of the new
drug candidate.
2. RA and QA personal: These members are to ensure that
the developmental studies meet with the regulatory
requirements and to see that the studies are conducted
according to regulatory guidelines.
3. Clinical researchers: They provide input into study designs
so that the generated results support the proposed clinical
program. They initiate development of protocols for clinical
safety, tolerance and efficacy as well as the investigators
brochure.
4. Analytical chemist: They are responsible for development
of assay methods and for characterize the new drug
substance and new drug product.
5. Manufacturing chemist: They scale up the synthesis of
the drug candidate and provide GLP/GMP quality material for
research studies.
6. Marketing experts: They help to determine drug
candidate's potential market through survey for other
companies drugs under development or drugs that are
already on the market for the disease indication.
Clinical Drug Development Team (CDDT) :
Once the IND is submitted, the non-clinical project team is
further expanded to include new members and renamed as
Clinical Drug Development Team (CDDT) wherein teams roles
substantially expand. Some of the old members, such as from
chemistry and pharmacology disciplines, have decreased roles
but may continue to serve on the team to provide scientific
expertise if new problems arise. The new players and their
expanded roles include:
1.Physicians: They are involved in conducting Phase I, II and
III clinical studies or serve as medical monitors if these studies
are conducted by a Clinical Research Organization (CRO).
2. Clinical research associates: They co- ordinate and
monitor the Phase I, II and III studies and ensure that the case
report forms (CRF) are correctly prepared.
3. Manufacturing chemist: They co- ordinate development of the
new drug substance and drug product production facilities and
ensure that all the necessary processes are in place.
4. Quality control analyst: They ensure that the appropriate assays
are developed, validated, and are in place to support the
manufacturing schedule.
5. Statisticians: Their responsibility is to assist in designing clinical
protocols and evaluate generated results from non-clinical and
clinical studies.
6. Clinical pharmacokinetics: They participate in Phase I trials to
support design of studies and conduct pharmacokinetic studies.
7. Marketing personal: They evaluate the status of competitor's
drug candidates and the potential of the new drug candidate to fulfill
a medical need and to prepare for product launch.
The responsibilities of these project teams
Non- Clinical Drug Development:
Pre- Clinical Trial: A laboratory test for a novel drug or a new medical device is
usually done on animal subjects, to see if the hoped for treatment really works and if
it is safe to test on humans.
The primary aims of the non-clinical (or pre clinical)
development phase is to analyze and determine which
candidate has the greatest probability of success, assess its
safety, and raise firm scientific foundations before transition to
the clinical development phase. This process of non clinical
development of medicine is very complex, time consuming
and regulatory driven.
The selected candidate compound should also meet non-
medical objectives, Which also include defining the IPR and
making enough medicinal products available for clinical trials.
Once identification of candidate compound is completed, the
non-clinical development should start answering the following
questions
Answers will come from specific assessments/studies:
Does it work? ---- Assessment of Efficacy.
How will it be delivered and how will the body react? ----
Profiling.
Is it safe? --- Toxicology/safety.
Is the manufacture viable and controllable? Non-clinical
development activities can continue throughout the lifecycle of
the product.
Various Pharmacological Approaches to Drug Discovery:
As an academic principle Pharmacology can be loosely
defend as the study of effects of chemical substances on
living systems. This definition is so broad that it holds all the
aspects of drug discovery, ranging from details of interaction
between drug molecule and its target to consequences of
placing the drug in the market.
Selectivity Testing: It consists of two main stages i.e.
screening for selectivity and Binding assay. To determine the
potency of drug, the selectivity of a compound for chosen
molecular target needs to be assessed.
Pharmacological Profiling: This includes the determination
of pharmacodynamics effect of new compound, either on In-
vitro models (which include cell lines or isolated tissues) or in-
vivo models (which include normal animals, animal models of
disease).
The aim of Pharmacological profiling is to answer the following
questions:
Do the molecular and cellular effects measured in screening assays
actually give rise to the predicted pharmacological effects in intact
tissues and whole animals?
Does the compound produce effects in intact tissues or whole
animals not associated with actions on its principle molecular target?
Is there a correspondence between potency of the drug at molecular
level, time level and the whole animal level? #
Does in-vivo duration of action match up with the pharmacokinetic
properties of the drug?
What happens if the drug is continuously or repeatedly given to an
animal over a course of days or weeks?
Does it loose its effectiveness or reveal effects not seen on acute
administration and whether there is any rebound after effect when it
is stopped.
SAFETY PHARMACOLOGY:
• This includes the scientific evaluation and study of potentially life
threatening pharmacological effects of a potential drug which is
unrelated to the desired therapeutic effect and therefore may
present a hazard.
• These tests are conducted at doses not too much in excess of
the intended clinical dose.
• Safety pharmacology seeks to identify unanticipated effects of
new drugs on major organ function (i.e. secondary
pharmacological effects).
• It is aimed at detecting possible undesirable or dangerous effects
of exposure of the drug in therapeutic doses.
• The emphasis is on acute effects produced by single-dose
administration rather than effects on chronic exposure as in
toxicological studies.
TOXICOLOGICAL APPROACHES TO DRUG DISCOVERY:
1. Acute Toxicity:
Studies should be carried out in at least two species, usually
mice and rats using the same route as intended for humans.
In addition, at least two more routes should be used to ensure
systemic absorption of the drug; this route may depend on the
nature of the drug. Mortality should be looked for up to 72
hours after parenteral administration and up to 7 days after
oral administration. The symptoms, signs and mode of death
should be reported, with appropriate macroscopic and
microscopic findings where necessary.
2. Long Term Toxicity:
These studies should be carried out in at least two mammalian
species and out of these two mammalian species one should be a
non rodent.
The duration of study will depend on the factor that whether the
application is for marketing permission or for clinical trial, and in the
later case, on the phases of trials.
If a species is known to metabolize the drug in the same way as
humans, it should be preferred in long-term toxicity studies. The
drug should be administered 7 days a week by the route intended
for clinical use in humans.
A control group of animals, given the vehicle alone, should always
be included, and three other groups should be given graded doses
of the drug; the highest dose should produce observable toxicity,
the lowest dose should not cause observable toxicity, but should be
comparable to the intended therapeutic dose in humans or multiple
of it.
Phase 0
Pharmacodynamics and pharmacokinetics in human- phase 0 trials
are optional first in human trials singles of therapeutic doses of the
study drug or treatment are given to a small number of subjects (
typically 10 to 15) together preliminary data pharmacodynamics and
pharmacokinetics parameters
Phase I
Trials
Screening for safety phase one consists of usually in health
valentines determine safety and dosing ( testing within a small group
of people typically 20-80 )
Phase II
Trials
Establishing the preliminary efficacy of drug usually against a
placebo - fresh to is used to get an initial grading of efficacy and
father explore safety in small number of patients having the disease
targeted by the NCE ( testing with large group of people typically 100-
300 )
Phase III
Trials
Final confirmation of safety and efficacy - phase 3 is large pivotal
trials to determine safety and efficacy in sufficiently large number of
patients with targeted disease. if safety and efficacy are adequately
proved, clinical testing may stop at the step at the NCE advances to
the new drug application ( NDA) stage ( testing with large group of
people typically 1000 to 3000 )
Phase IV
Trials
Safety studies during sales - phase 4 is post approval trials that are
sometimes a condition attached by the FDA, also called post market
surveillance studies.
Name of the Application For What
IND (Investigational new drug) It is an application filled to the FDA in
order to start clinical trials in humans on
the basis of the data obtained from the
printer clinical trials.
NDA ( new drug application) If clinical studies confirm that a new
drug is relatively safe and effective and
will not pose a reasonable risks to
patients.
ANDA ( abbreviated new drug
application )
It is made for approval of genetic drugs.
BLA ( biologic license application ) Biology products are approved for
marketing under the provisions of the
public health service act.
Investigational New Drug (IND) Application:
Investigational New Drug (IND) is a program by which any
Pharmaceutical company can approach to obtain permission
for the initiation of human clinical trials and an experimental
drug across state lines (usually to clinical investigators) before
a marketing application for the drug has been approved.
An investigational new drug (IND) application is to provide the
data showing that it is reasonable to begin tests of a new drug
on humans.
The IND application is also the vehicle through which a
sponsor advances to the next stage of drug development
known as clinical trials.
Current Federal law requires that a drug be the subject of an
approved marketing application before it is transported or
distributed across state lines (Clinical Investigators).
Because a sponsor will probably want to ship the investigational drug to
clinical investigators in many states, it must seek an exemption from that
legal requirement.
The IND application is the means through which the sponsor technically
obtains this exemption from the FDA
During a new drug's early preclinical development, the sponsor's primary
goal is to determine if the product is reasonably safe for initial use in
humans and if the compound exhibits pharmacological activity that justifies
commercial development.
When a product is identified as a viable candidate for further development,
the sponsor then focuses on collecting the data and information necessary
to establish that the product will not expose humans to unreasonable risks
when used in limited, early-stage clinical studies.
FDA‘s role in the development of a new drug begins when the drug's
sponsor (usually the manufacturer or potential marketer), having screened
the new molecule for pharmacological activity and acute toxicity potential in
animals, wants to test its diagnostic or therapeutic potential in humans.
At that point, the molecule changes in legal status under the
Federal Food, Drug, and Cosmetic Act and becomes new drug
subject to specific requirements of the drug regulatory system.
Types of IND Applications:
1. Investigator IND Application,
2. Emergency use IND application,
3. Treatment IND application,
4. Screening IND application.
1.Investigation IND application: In this an application is submitted
by a physician, who both initiates and conducts an investigation, and
under whose immediate direction the investigational drug is
administered or dispensed.
A physician might submit a research IND application to propose
studying of:
An unapproved drug,
An approved product for a new indication or
An approved product in a new patient population.
2. Emergency use IND Application: This application allows the FDA to authorize
use of an experimental drug in an emergency situation that does not allow time for
submission of an IND application, in accordance with 21CFR, Sec. 312.23 or Sec.
312.20. It is also used for patients who do not meet the criteria of an existing study
protocol, or if an approved study protocol does not exist. In such a case, FDA may
authorize shipment of the drug for a specified use in advance of submission of an
IND application.
3. Treatment IND Application: This application is submitted for experimental drugs
showing promise in clinical testing for serious or immediately life-threatening
conditions while the final clinical work is conducted and the FDA review takes place.
A drug that is not approved for marketing may be under clinical investigation for a
serious or immediately life- threatening disease condition in patients for whom no
comparable or satisfactory alternative drug or other therapy is available.
In the case of a serious disease, a drug ordinarily may be made available for
treatment use during phase III investigations or after all clinical trials have been
completed.
In the case of an immediately life-threatening disease a drug may be made available
for treatment use earlier than phase III but ordinarily not earlier than phase II.
4. Screening IND Application: It is flied for multiple,
closely related compounds in order to screen for the
preferred compounds or formulations. The preferred
compound can be developed under a separate IND. It can
also be used for screening different salts, esters and other
drug derivatives that are chemically different, but
pharmacodynamically similar.
IND Review and Report: During this time, FDA has an
opportunity to review the IND application for safety to
assure that research subjects will not be subjected for
unreasonable risk. The report evaluates on the various
factors like Medical Review, Chemistry review,
Pharmacology/Toxicology review, Statistical analysis and
Safety review.
Layout chart for IND Application
Investigators Brochure (IB):- The Investigator‘s Brochure (IB) is a
compilation of the clinical and non-clinical data on the investigational
product(s) that are relevant to the study of the product(s) in human
subject.
1.Purpose of Investigator‘s Brochure (IB) : Its purpose is to
provide information to the investigators and others involved in the
trial such as the dose, dose frequency/interval, methods of
administration and safety monitoring procedures.
The IB also provides insight to support the clinical management of
the study Subjects during the course of the clinical trial. The
information should be presented in a concise and simple manner.
IB enables a clinician, or potential investigator, to understand it and
make his/her own unbiased ( without any partiality) risk-benefit
assessment of the appropriateness of the promised trial. For this
reason a medically qualified person should generally participate in
the editing of an IB.
2. Contents of Investigators Brochure :
1.Table of contents.
2. Summary not exceeding 2 pages, highlighting the significant physical,
chemical, Pharmaceutical, Pharmacological, toxicological,
pharmacokinetic, metabolic, and clinical information available of IP.
3. Introduction: Chemical name, active ingredients, pharmacological
class, anticipated therapeutic/diagnostic indication(s). General approach to
be followed in evaluating the IP.
4. Description as per IP: Physical, chemical and pharmaceutical
properties as per IP storage and handling as per IP. Any structural
similarity with the other known compound given.
5. Non-Clinical Studies: The results of all relevant non- clinical
pharmacology, toxicology, pharmacokinetic, and investigational product
metabolism studies should be provided in summary form.
The information provided may include: Species tested, Unit dose (e.g.,
milligram/kilogram (mg/kg), Dose interval, Route of administration and
Duration of dosing.
5.1 Non-Clinical Pharmacology: A summary of the
pharmacological aspects of the investigational product studied
in animals should be included.
5.2 Pharmacokinetics and Product Metabolism in Animals:
A summary of the pharmacokinetics (ADME) and biological
transformation and disposition (getting a drug into its
appropriate position in the body and in an appropriate
concentration) of the investigational product in all species
studied should be give
5.3 Toxicology: (The study of the adverse effects of chemicals
on animals): A summary of the toxicological effects found in
relevant studies conducted in different animal species. (Single
dose, Repeated dose, Carcinogenicity, Special studies
(irritancy, sensitization), Reproductive toxicity).
6. Effects in Humans: A thorough discussion of the known
effects of the investigational product(s) in humans should be
provided, including information on Pharmacokinetics,
metabolism, Pharmacodynamics, dose response, safety,
efficacy, and other pharmacological activities like
Pharmacokinetics and Product Metabolism in Humans
7. Summary of Data and Guidance for the investigator:
This section should contain non-clinical and clinical data of IP.
Investigators Brochure (IB) provides the Investigator a clear
understanding of the possible risks, adverse reactions,
observations and precautions needed for the clinical trial.
NEW DRUG APPLICATION (NDA)
Definition: An NDA is an application to permit the sale and marketing of a
new drug. The vehicle through which drug sponsors formally propose that
the regulatory body approves a new pharmaceutical products for sale and
marketing, and the data gathered during the animal studies and human
clinical trials of an investigational new product becomes a part of the NDA.
Aim of NDA: The aims of NDA include providing adequate information to
permit FDA reviewers to establish the following:
 Safety and effectiveness of drug,
 Benefits overweigh risks,
 Is the drug's proposed labeling (package insert) appropriate, and what
should it contain?
 Are the methods used in manufacturing (Good Manufacturing Practice,
GMP) the drug and the controls used to maintain the drug's quality
adequate to preserve the drug's identity, strength, quality, and purity?
Risk Benefit.
NDA Contents
NDA Review Process:
Once the application is submitted, the FDA has 60 days to
conduct a preliminary review which will assess whether the
NDA is "sufficiently complete to permit a substantive review".
If everything is found to be acceptable, the FDA will decide if
the NDA will get a standard or accelerated review and
communicate the acceptance of the application and their
review choice in another communication known as the 74-
day letter
NDA Review Process
Bio Equivalence Studies:
BE studies are very essential to ensure uniformity in
standards of quality, efficacy and safety of pharmaceutical
products so that reasonable assurance can be provide for the
various products containing same active ingredient, marketed
by different licensees are clinically equivalent and
interchangeable.
Both Bioavailability and Bioequivalence focus on release of
drug substance from its dosage form and subsequent
absorption in circulation.
Similar approaches to measure bioavailability should be
followed in demonstrating bioequivalence.
Types of Equivalences
Bioavailability:
Bioavailability means 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.
Equivalence:
It is a relative term that compares drug products with respect
to a specific characteristic or function or to a defined set of
standards.
Clinical Research Protocols:
It is a complete written description of and scientific rationale for a
research activity involving human subjects.
Sufficient information is to be gathered on the quality of the non-
clinical safety to conduct the protocol and health authority/ethics
committee approval is granted in the country where approval of the
drug or device is sought (i.e. in past).
The clinical trial design and objectives are written into a document
called a clinical trial protocol. It is a document that states the
background, objectives, rationale, design, methodology (including
the methods for dealing with AEs i.e. Adverse Drug Events,
withdrawals etc.) and statistical considerations of the study. It also
states the conditions under which the study shall be performed and
Look for better ways to prevent disease in people who never had the
disease or to prevent a disease from returning.
The protocols means:
To clarify the research question.
To compile existing knowledge.
To formulate a hypothesis and objectives.
To decide about a study design.
To clarify ethical considerations.
To apply for funding.
To have a guideline and tool for the research team.
Parts of the Protocol:
1.Title Page.
2. Signature Page.
3. Content Page.
4. List of Abbreviations.
5. Introduction/Abstract.
6. Objectives.
7. Background/Rationale.
8. Eligibility Criteria .
9. Study Design/Methods (Including Drug/Device Info).
10. Safety/Adverse Events.
11. Regulatory Guidance.
12. Statistical Section (Including Analysis and Monitoring).
13. Human Subjects Protection/Informed Consent.
Biostatistics in Pharmaceutical Product Development:
Biostatistics is a science where professionals collect, analyze
and interpret conclusions for data. In simpler words, when we
apply statistics in biological science it is called biostatistics.
It requires an understanding of statistical theory and
application to work out issues within biological and health
science organizations.
Biostatisticians typically address healthcare topics, either in
the private or public sectors, and work in an office
environment.
The role of the biostatistician in pharmaceutical product
development is more important than ever, and hence statistics
professionals need to develop better approaches to solve
some old limitations and some new problems
Pharmaceutical biostatisticians analyze genetic data and
disease rates. They use this information to design clinical trials
to evaluate new drugs.
They study populations exposed to harmful chemicals to
determine their overall effects.
For example, during a clinical trial, there are many phases for
drug or device development. To check the outcome of the
study as trial is going good or bad.
biostatisticians applies concepts of statistics and do the
analysis. Based on the analysis results, they draw inference
about the study. So biostatistics is a method that comprises of
a collection of data, summarizing, analyzing and drawing
inferences.
Basic Concepts of Biostatistics:
1.Statistical Levels and Decision Making:
Hypothesis testing is a method used to determine whether the
results are statistically significant or not.
It is the process of drawing inferences to make decision about
the sample with regards to the population as a whole.
Bio statistical analyses in human clinical trials are required
during the last stages of the pharmaceutical product
development process. It has effectively become a hurdle for
every new drug, device, and biological while on the way to
market.
2. Sample Size: Sampling is a method used in statistical analysis of a data
where a specific number of samples are taken from a population for a study.
For example, health ministry wants to know the opinion or suggestions of
recently implemented health program in particular region.
Here the population is that region and sample is cities in that region. In the
demonstration of efficacy of a new product, achieving p-value ≤ 0.05 is critical
while comparing the new product to the control group.
It is observed that most products with modest potential therapeutic benefit
complies this specification only if the sponsor of the product invest enough
money and/or time to conduct a very large trial.
Sample size can overcome the limitations of such benefits and hence is the true
decision criteria for effectiveness.
Use of huge financial investment is not the best model for the evaluation and
approval of new products.
Product approval should not be based on the financial strength of the sponsor
and/or product's future revenue potential. Rather, safety concerns of clinical trial
participants and the future subjects of the product are the important elements.
Thus, sound statistical criteria should be used to quantify sample size and to
ensure public health concerns
3 Data Collection: it is first thing in any kind of analysis. Data are the
actual pieces of information that we collect through experiments. It
can be categorized into categorical and numerical data form.
Categorical data are defined in categories or groups. For example,
male or female, yes or no, etc.
Numerical data are classified into discrete and continuous data.
Discrete data are the data which take certain values. For example,
number of patients, number of devices etc.
Continuous data are the data which can take any values either
fractional or decimal. For example, blood pressure level, height and
weight of the patient etc.
Data collection in a clinical trial begins as clinical information is
collected from subjects. Initially it reflects some component of truth
about treatment effects, and some component of variability or error.
Statistical analysis techniques can estimate the magnitude of the
error component. This must be used as a metric to estimate the size
and reliability of the truth component.
Biostatisticians generally require consistent and relatively simple
clinical assessments to create data tabulations and perform analyses.
As they are engaged in data cleaning raw data often does not meet
this expectation and thus resulting altered data consists of truth, error
and systematic bias. The impact of systematic bias introduced during
the cleaning process is not normally assessed statistically.
4. Measurement System Analysis: Measurement system analysis
helps to detect the amount of variation existing within a measurement
system. Suppose during the initial clinical trial phase of a particular
product, if the data are not recorded properly, and if further analysis is
to be carried out then consequences of it would be a failure at final
phases.
The intention of clinical trials is to accurately predict benefits and risks
of new drug product to the broad population of subjects, while
protecting the safety of those subjects who volunteer to provide the
data needed to make that prediction.
Experimental Design in Clinical Trials:
As seen under clinical trials section, these trials for a new drug,
device, or biological typically are conducted from Phase-l to Phase-3
prior to NDA submission to a regulatory agency for approval.
Each phase involves increase in number of subjects sequentially.
Biostatistician is involved in the design and analysis of these studies.
Biostatistician in collaboration with medical, regulatory and data
management experts defines what to measure, how often to measure
it, how to select subjects and randomly assign treatments to them,
and how to analyze the outcome.
All these things are pre-specified in the clinical protocol, including the
expected outcome, all analysis strategies, and the rationale for the
number of subjects to be incorporated in the studies.
1.Sample Size: In order to establish the sample size, a statistician is
consulted at the beginning of a trial. Deciding required sample size is critical
for Phase-3 trials.
This is because statistical significance in the final analyses at this phase is
required to support a submission for MAA for approval. Insufficient sample
size may lead to failures for drugs to appear as adequately safe and
effective for approval. The outcome of Phase-2 trial provides enough
evidence to establish the required sample size for Phase-3 trials.
The overall results from sample reflect both overall population response and
some degree of random variability. The larger the sample size, the smaller
the effect of the variability on the overall results.
Inferential statistics helps to quantify uncertainty in effect of the variability.
The statistical power measures the degree to which the proposed sample
size can overcome unpredictable variability in subject response, as well as
the degree of confidence that the results of the sample will approximate the
future response of the population.
2. Sensitivity and Cost: The increase in the sample size in a
clinical trials increases the level of assurance that the drug is an
effective drug and ultimately the statistical significance when
the results are analyzed using statistical methods.
As sponsor has some financial and time constraint, sample
sizes are always a compromise between the ideal and the
affordable. This is crucial stage where a biostatistician is
involved to define the parameters for sample size.
The sensitivity of a trial to detect a statistically significant value
also depends on sample size. Since all other things are almost
equal, the larger the sample size, the less effective a drug must
be to show a significant, and potentially approvable, result.
Financial investment in clinical research is an important factor
for obtaining marketing approval.
3. Random Sampling:
The random sampling in clinical trials has two major
requirements for inferential statistics to forecast population
benefits.
First, it is necessary that the subjects in clinical trials must be
randomly selected and it must be a representative sample of
the population of future subjects in the total population.
Second, it is essential that for comparative trials, treatments
must be randomly assigned to subjects in clinical trial.
Commonly major trials follows later requirement, and in some
trials the earlier requirement is complied.
4. Data Refinement: The CRFs are developed to increase clinical
database to fulfill the requirements of the statistician for analysis and
reporting and the sponsor to establish sufficient evidence for approval.
The recommendations to avoid bias in data refinement and categorization
activities are given below:
(i) Clinical trial data captured during study should be categorized into its
objectivity or subjectivity, immediacy or delay of collection and source.
(ii) Apply objective data cleaning criteria, for example, range checks and
consistency comparisons.
(iii) For subjective data exercise extreme caution when questioning values
(especially when a time has elapsed since its collection).
(iv) Formalized coding schemes should be relevant to the drug under study.
(v) Data cleaning efforts for variables that are not Identified as critical
should be minimized.
DATA PRESENTATION FOR FDA SUBMISSIONS: Study data standards
describe a standard way to exchange clinical and non-clinical study data.
These standards provide a consistent general framework for organizing
study data, including templates for datasets, standard names for variables;
identify appropriate controlled terminology and standard ways of doing
calculations with common variables.
Data standards also help FDA receive, process, review, and archive
submissions more efficiently and effectively.
FDA has been working towards a standardized approach to capture,
receive and analyze study data.
Standardization of study data is vital to integrate pre- marketing study data
and post-marketing safety data to improve public health and patient safety.
Central to this vision is the creation of an enterprise data infrastructure
(Janus) within FDA to improve the management of all structured scientific
data.
Data Standards: Data standards can be divided into two
categories:
1. Exchange Standards: Exchange standards provide a
consistent way to exchange information. Exchange standards
help to ensure that the sending and the receiving systems
both understand unambiguously what information is being
exchanged.
2. Terminology Standards: Terminology standards provide a
consistent way to describe concepts. For example, the Unique
Ingredient Identifiers (UNII), developed by the FDA, provides a
consistent way to describe substances in foods and drugs.
Vocabulary developed National Institute of Cancer describes
terminology related to cancer.
MANAGEMENT OF CLINICAL STUDIES:
Clinical trial management is most simply defined as the
process that an organization follows to ensure that quality
(defined as minimized risks and clean data) is delivered
efficiently and punctually.
It refers to a standards-driven process that a project manager
initiates and follows in order to successfully manage clinical
trial sites, clinical research associates, and workflow by using
clinical trial management tools or software prolonged timelines
and heavy costs related to large trials have been prompted a
new focus on more eminent clinical trial management.
It is possible to dramatically reduce the total cost of a clinical
trial by 60% 90% without compromising the scientific validity of
the results.
Life Cycle of Clinical Trial Project: A more accurate control, regardless of
the therapeutic area or trial stages (all the way through from 'preclinical’
phase 1 to 'post- approval' phase 4 studies), is ensured by typically
breaking down the life cycle of each clinical trial project into 4 phases:
Conceptual, Planning, Implementation and Analysis.
Clinical Trial Protocol: A protocol is a document that describes the
purpose, design, methodology, statistical considerations and organization of
a study, and provides basic information and rationale for the clinical study.
The contents that should be present in the protocol are described by the
GCP. The protocol writing is a task for one person, usually the principal
investigator, not a committee.
There are various challenges of Project management in clinical trials.
Clinical trials all need the same coordinated processes and systems,
irrespective of the size, scope, costs, or period. The key challenge is then to
implement and maintain effective management systems and techniques in
response to the needs of the trial project.
Thank you

More Related Content

What's hot

Technology transfer.pptx
Technology transfer.pptxTechnology transfer.pptx
Technology transfer.pptxAshwiniBhoir2
 
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.GvDurgamani
 
Pharmaceutical Licecnsing authorites of india
Pharmaceutical Licecnsing authorites of indiaPharmaceutical Licecnsing authorites of india
Pharmaceutical Licecnsing authorites of indiaAtul Bhombe
 
Technology Transfer Related Documents.pptx
Technology Transfer Related Documents.pptxTechnology Transfer Related Documents.pptx
Technology Transfer Related Documents.pptxAfroj Shaikh
 
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...Audumbar Mali
 
Regulatory requirements for drug approval - industrial pharmacy II
Regulatory requirements for drug approval - industrial pharmacy IIRegulatory requirements for drug approval - industrial pharmacy II
Regulatory requirements for drug approval - industrial pharmacy IIJafarali Masi
 
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCT
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCTCopp - CERTIFICATE OF PHARMACEUTICAL PRODUCT
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCTSuraj Pamadi
 
Regulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIRegulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIJafarali Masi
 
Regulatory Requirements For New Drug Approval
Regulatory Requirements For New Drug ApprovalRegulatory Requirements For New Drug Approval
Regulatory Requirements For New Drug ApprovalShagufta Farooqui
 
Patients medication history interview
Patients medication history interviewPatients medication history interview
Patients medication history interviewSubhash Yende
 
Implantable Drug Delivery System
Implantable Drug Delivery SystemImplantable Drug Delivery System
Implantable Drug Delivery SystemSourav Kar
 
Regulatory affairs overview.pptx
Regulatory affairs overview.pptxRegulatory affairs overview.pptx
Regulatory affairs overview.pptxkajal pradhan
 
Indian Regulatory Requirements-1.pptx
Indian Regulatory Requirements-1.pptxIndian Regulatory Requirements-1.pptx
Indian Regulatory Requirements-1.pptxTejasBhatia2
 
Investigational use of drugs
Investigational use of drugsInvestigational use of drugs
Investigational use of drugssunayanamali
 

What's hot (20)

Technology transfer.pptx
Technology transfer.pptxTechnology transfer.pptx
Technology transfer.pptx
 
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.
WHO GUIDELINES FOR TECH.TRANSFER SIDHANTA SAHU.
 
Pharmaceutical Licecnsing authorites of india
Pharmaceutical Licecnsing authorites of indiaPharmaceutical Licecnsing authorites of india
Pharmaceutical Licecnsing authorites of india
 
Technology Transfer Related Documents.pptx
Technology Transfer Related Documents.pptxTechnology Transfer Related Documents.pptx
Technology Transfer Related Documents.pptx
 
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...
Industrial Pharmacy-II (IP-II) Unit 2:- chapter:- 2 Technology Development an...
 
Regulatory requirements for drug approval - industrial pharmacy II
Regulatory requirements for drug approval - industrial pharmacy IIRegulatory requirements for drug approval - industrial pharmacy II
Regulatory requirements for drug approval - industrial pharmacy II
 
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCT
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCTCopp - CERTIFICATE OF PHARMACEUTICAL PRODUCT
Copp - CERTIFICATE OF PHARMACEUTICAL PRODUCT
 
Regulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy IIRegulatory affairs - industrial pharmacy II
Regulatory affairs - industrial pharmacy II
 
Supac
Supac Supac
Supac
 
Regulatory Requirements For New Drug Approval
Regulatory Requirements For New Drug ApprovalRegulatory Requirements For New Drug Approval
Regulatory Requirements For New Drug Approval
 
New Drug Approval in India
New Drug Approval in IndiaNew Drug Approval in India
New Drug Approval in India
 
Patients medication history interview
Patients medication history interviewPatients medication history interview
Patients medication history interview
 
Implantable Drug Delivery System
Implantable Drug Delivery SystemImplantable Drug Delivery System
Implantable Drug Delivery System
 
COPP.pptx
COPP.pptxCOPP.pptx
COPP.pptx
 
Granularity of TT Process.pdf
Granularity of TT Process.pdfGranularity of TT Process.pdf
Granularity of TT Process.pdf
 
Regulatory affairs overview.pptx
Regulatory affairs overview.pptxRegulatory affairs overview.pptx
Regulatory affairs overview.pptx
 
Indian Regulatory Requirements-1.pptx
Indian Regulatory Requirements-1.pptxIndian Regulatory Requirements-1.pptx
Indian Regulatory Requirements-1.pptx
 
Investigational use of drugs
Investigational use of drugsInvestigational use of drugs
Investigational use of drugs
 
Regulatory Affairs Profession
Regulatory Affairs ProfessionRegulatory Affairs Profession
Regulatory Affairs Profession
 
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATIONCDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
CDSCO- CENTRAL DRUG STANDARD CONTROL ORGANISATION
 

Similar to regulatory requirements for drug approval ( IP-2 / UNIT -3 )

Regulatory requirements for drug approval
Regulatory requirements for drug approvalRegulatory requirements for drug approval
Regulatory requirements for drug approvalAudumbar Mali
 
CLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxCLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxTejaswiniAsawa
 
Various process of drug development
Various process of drug developmentVarious process of drug development
Various process of drug developmentAyanpal33
 
Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,PakistanPharmaCareerDoor
 
Drug development team
Drug development teamDrug development team
Drug development teamChandrikaSundru
 
The stages of Drug Discovery and Development process
The stages of Drug Discovery and Development processThe stages of Drug Discovery and Development process
The stages of Drug Discovery and Development processA M O L D E O R E
 
Drug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsDrug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsHeena Parveen
 
Drug development process
Drug development process Drug development process
Drug development process Zobayer Hossain
 
Development of drug
Development of drugDevelopment of drug
Development of drugHind Alshankiti
 
Stages of drug development
Stages of drug developmentStages of drug development
Stages of drug developmentmeethy
 
Features of clinical trials
Features of clinical trialsFeatures of clinical trials
Features of clinical trialsDRASHTI PATEL
 
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial_Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLSKatalyst HLS
 
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSKatalyst HLS
 
Introduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxIntroduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxMingmaLhamuBhutia
 
Drug development process
Drug development processDrug development process
Drug development processSailesh Mahapatra
 
Drug Discovery and Development.pptx
Drug Discovery and Development.pptxDrug Discovery and Development.pptx
Drug Discovery and Development.pptxVitthal Mane
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development ProcessTusharJ7
 
Application of bio-pharmaceutics in new drug development .
Application of bio-pharmaceutics in new drug development .Application of bio-pharmaceutics in new drug development .
Application of bio-pharmaceutics in new drug development .MD SAYDUR RAHMAN
 
Non-clinical drug development
Non-clinical drug developmentNon-clinical drug development
Non-clinical drug developmentJayeshRajput7
 
Non clinical drug development
Non clinical drug developmentNon clinical drug development
Non clinical drug developmentAbhimanyu Awasthi
 

Similar to regulatory requirements for drug approval ( IP-2 / UNIT -3 ) (20)

Regulatory requirements for drug approval
Regulatory requirements for drug approvalRegulatory requirements for drug approval
Regulatory requirements for drug approval
 
CLINICAL TRIALS.pptx
CLINICAL TRIALS.pptxCLINICAL TRIALS.pptx
CLINICAL TRIALS.pptx
 
Various process of drug development
Various process of drug developmentVarious process of drug development
Various process of drug development
 
Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,Lecture # 03 New Drug Approval and Development Process,
Lecture # 03 New Drug Approval and Development Process,
 
Drug development team
Drug development teamDrug development team
Drug development team
 
The stages of Drug Discovery and Development process
The stages of Drug Discovery and Development processThe stages of Drug Discovery and Development process
The stages of Drug Discovery and Development process
 
Drug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugsDrug discovery & clinical evaluation of new drugs
Drug discovery & clinical evaluation of new drugs
 
Drug development process
Drug development process Drug development process
Drug development process
 
Development of drug
Development of drugDevelopment of drug
Development of drug
 
Stages of drug development
Stages of drug developmentStages of drug development
Stages of drug development
 
Features of clinical trials
Features of clinical trialsFeatures of clinical trials
Features of clinical trials
 
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial_Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial_Katalyst HLS
 
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLSDiscovery of Drug and Introduction to Clinical Trial__Katalyst HLS
Discovery of Drug and Introduction to Clinical Trial__Katalyst HLS
 
Introduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptxIntroduction to drug discovery and development.pptx
Introduction to drug discovery and development.pptx
 
Drug development process
Drug development processDrug development process
Drug development process
 
Drug Discovery and Development.pptx
Drug Discovery and Development.pptxDrug Discovery and Development.pptx
Drug Discovery and Development.pptx
 
Drug Development Process
Drug Development ProcessDrug Development Process
Drug Development Process
 
Application of bio-pharmaceutics in new drug development .
Application of bio-pharmaceutics in new drug development .Application of bio-pharmaceutics in new drug development .
Application of bio-pharmaceutics in new drug development .
 
Non-clinical drug development
Non-clinical drug developmentNon-clinical drug development
Non-clinical drug development
 
Non clinical drug development
Non clinical drug developmentNon clinical drug development
Non clinical drug development
 

Recently uploaded

Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Celine George
 
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxiammrhaywood
 
Meghan Sutherland In Media Res Media Component
Meghan Sutherland In Media Res Media ComponentMeghan Sutherland In Media Res Media Component
Meghan Sutherland In Media Res Media ComponentInMediaRes1
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxRaymartEstabillo3
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...Marc Dusseiller Dusjagr
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxthorishapillay1
 
Painted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaPainted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaVirag Sontakke
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxOH TEIK BIN
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceSamikshaHamane
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxmanuelaromero2013
 
Earth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatEarth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatYousafMalik24
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfMahmoud M. Sallam
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Celine George
 
Hierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementHierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementmkooblal
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPCeline George
 
Historical philosophical, theoretical, and legal foundations of special and i...
Historical philosophical, theoretical, and legal foundations of special and i...Historical philosophical, theoretical, and legal foundations of special and i...
Historical philosophical, theoretical, and legal foundations of special and i...jaredbarbolino94
 
ESSENTIAL of (CS/IT/IS) class 06 (database)
ESSENTIAL of (CS/IT/IS) class 06 (database)ESSENTIAL of (CS/IT/IS) class 06 (database)
ESSENTIAL of (CS/IT/IS) class 06 (database)Dr. Mazin Mohamed alkathiri
 
Final demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxFinal demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxAvyJaneVismanos
 

Recently uploaded (20)

Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17Computed Fields and api Depends in the Odoo 17
Computed Fields and api Depends in the Odoo 17
 
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptxECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
ECONOMIC CONTEXT - PAPER 1 Q3: NEWSPAPERS.pptx
 
Meghan Sutherland In Media Res Media Component
Meghan Sutherland In Media Res Media ComponentMeghan Sutherland In Media Res Media Component
Meghan Sutherland In Media Res Media Component
 
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptxEPANDING THE CONTENT OF AN OUTLINE using notes.pptx
EPANDING THE CONTENT OF AN OUTLINE using notes.pptx
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
 
Proudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptxProudly South Africa powerpoint Thorisha.pptx
Proudly South Africa powerpoint Thorisha.pptx
 
Painted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of IndiaPainted Grey Ware.pptx, PGW Culture of India
Painted Grey Ware.pptx, PGW Culture of India
 
Solving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptxSolving Puzzles Benefits Everyone (English).pptx
Solving Puzzles Benefits Everyone (English).pptx
 
Roles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in PharmacovigilanceRoles & Responsibilities in Pharmacovigilance
Roles & Responsibilities in Pharmacovigilance
 
How to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptxHow to Make a Pirate ship Primary Education.pptx
How to Make a Pirate ship Primary Education.pptx
 
Earth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice greatEarth Day Presentation wow hello nice great
Earth Day Presentation wow hello nice great
 
Pharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdfPharmacognosy Flower 3. Compositae 2023.pdf
Pharmacognosy Flower 3. Compositae 2023.pdf
 
Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17Difference Between Search & Browse Methods in Odoo 17
Difference Between Search & Browse Methods in Odoo 17
 
Hierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of managementHierarchy of management that covers different levels of management
Hierarchy of management that covers different levels of management
 
What is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERPWhat is Model Inheritance in Odoo 17 ERP
What is Model Inheritance in Odoo 17 ERP
 
Historical philosophical, theoretical, and legal foundations of special and i...
Historical philosophical, theoretical, and legal foundations of special and i...Historical philosophical, theoretical, and legal foundations of special and i...
Historical philosophical, theoretical, and legal foundations of special and i...
 
OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...OS-operating systems- ch04 (Threads) ...
OS-operating systems- ch04 (Threads) ...
 
ESSENTIAL of (CS/IT/IS) class 06 (database)
ESSENTIAL of (CS/IT/IS) class 06 (database)ESSENTIAL of (CS/IT/IS) class 06 (database)
ESSENTIAL of (CS/IT/IS) class 06 (database)
 
Final demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptxFinal demo Grade 9 for demo Plan dessert.pptx
Final demo Grade 9 for demo Plan dessert.pptx
 

regulatory requirements for drug approval ( IP-2 / UNIT -3 )

  • 2. CONTENTS Introduction Drug development team Clinical drug development team Non-clinical drug development Various pharmacological approaches to drug discovery Safety Pharmacology Toxicological approaches to drug discovery IND Application ND Application Clinical Research protocols Basic concepts of Biostatistics Data presentation for FDA submissions Management of clinical studies
  • 3. INTRODUCTION Currently different nations have to follow different requirements for the regulatory approval of novice (novel) drug. It is almost difficult for every country to have the same regulatory approach for the Marketing Authorization Application (MAA). Therefore it is necessary to have knowledge about regulatory requirements for MAA of each country.
  • 4. Different phases of clinical trials
  • 5. New Drug Application (NDA) is an application submitted to the respective regulatory authority for permission to market a new drug. To obtain this permission a sponsor submits preclinical and clinical test data for analyzing the drug information and description of manufacturing procedures. After agency received the NDA, it undergoes a technical screening. This process of evaluation is made to ensure that the sufficient data and the required information have been submitted in each area justifying the "filling" application form.
  • 6. Possible action after the review of NDA
  • 7. Drug Development Team: Drug development is the process of bringing a new pharmaceutical drug to the market once a lead compound has been identified through the process of drug discovery. It includes pre-clinical research on microorganisms and animals, filing for regulatory status, for an investigational new drug (IND) to initiate clinical trials on humans, and may include the steps of obtaining regulatory approval with a new drug application to market the drug.
  • 8. The process of drug discovery and development is very long and needs 10-12 years which includes the close interaction of large number of scientific disciplines. Most Biotechnology and Pharmaceutical companies employ teams to mentor the process of various stages of drug development and making the drug candidate into therapeutic products
  • 9. The DDT is comprised of different sub-teams namely drug discovery team, preclinical drug development team and clinical drug development team. (a) Drug Discovery Team: Drug discovery team is a small group of researchers and is the first team in DDT. The primary responsibility of this team is to identify lead compounds or classes of compounds. They screen existing compounds in the company’s archives or in the libraries of compounds that are optimized for activity using combinatorial chemistry.
  • 10. The primary endpoint used in this assessment is the potency of the various lead compounds in the disease model. The pharmacology results are shared with the chemists, who then prepare analogues of the most active compounds to explore further the structure activity relationship (SAR). Once a lead compound or class of lead compounds has been identified, this team commonly grows to include other scientific disciplines to characterize more fully the possibility of developing the lead compounds of interest.
  • 11. Drug discovery team then includes other experts from various scientific disciplines such as pharmacology, chemistry, toxicology, drug metabolism and pharmacokinetics, and bio pharmaceutics. (b) Preclinical Drug Development Team: Once the management accepts a lead compound candidate as a drug candidate, its further establishment is carried out by preclinical drug development team (PDDT). This new team also involves researchers from the various scientific disciplines. Some companies have Separate groups to support discovery research and for conducting non-clinical and clinical developmental studies.
  • 12. In addition to the drug discovery team members, the PDDT has a number of new players that include: 1. A management: They assign team leader and coordinator to the team who responsible for the development of the new drug candidate. 2. RA and QA personal: These members are to ensure that the developmental studies meet with the regulatory requirements and to see that the studies are conducted according to regulatory guidelines. 3. Clinical researchers: They provide input into study designs so that the generated results support the proposed clinical program. They initiate development of protocols for clinical safety, tolerance and efficacy as well as the investigators brochure.
  • 13. 4. Analytical chemist: They are responsible for development of assay methods and for characterize the new drug substance and new drug product. 5. Manufacturing chemist: They scale up the synthesis of the drug candidate and provide GLP/GMP quality material for research studies. 6. Marketing experts: They help to determine drug candidate's potential market through survey for other companies drugs under development or drugs that are already on the market for the disease indication.
  • 14. Clinical Drug Development Team (CDDT) : Once the IND is submitted, the non-clinical project team is further expanded to include new members and renamed as Clinical Drug Development Team (CDDT) wherein teams roles substantially expand. Some of the old members, such as from chemistry and pharmacology disciplines, have decreased roles but may continue to serve on the team to provide scientific expertise if new problems arise. The new players and their expanded roles include: 1.Physicians: They are involved in conducting Phase I, II and III clinical studies or serve as medical monitors if these studies are conducted by a Clinical Research Organization (CRO). 2. Clinical research associates: They co- ordinate and monitor the Phase I, II and III studies and ensure that the case report forms (CRF) are correctly prepared.
  • 15. 3. Manufacturing chemist: They co- ordinate development of the new drug substance and drug product production facilities and ensure that all the necessary processes are in place. 4. Quality control analyst: They ensure that the appropriate assays are developed, validated, and are in place to support the manufacturing schedule. 5. Statisticians: Their responsibility is to assist in designing clinical protocols and evaluate generated results from non-clinical and clinical studies. 6. Clinical pharmacokinetics: They participate in Phase I trials to support design of studies and conduct pharmacokinetic studies. 7. Marketing personal: They evaluate the status of competitor's drug candidates and the potential of the new drug candidate to fulfill a medical need and to prepare for product launch.
  • 16. The responsibilities of these project teams
  • 17. Non- Clinical Drug Development: Pre- Clinical Trial: A laboratory test for a novel drug or a new medical device is usually done on animal subjects, to see if the hoped for treatment really works and if it is safe to test on humans.
  • 18. The primary aims of the non-clinical (or pre clinical) development phase is to analyze and determine which candidate has the greatest probability of success, assess its safety, and raise firm scientific foundations before transition to the clinical development phase. This process of non clinical development of medicine is very complex, time consuming and regulatory driven. The selected candidate compound should also meet non- medical objectives, Which also include defining the IPR and making enough medicinal products available for clinical trials. Once identification of candidate compound is completed, the non-clinical development should start answering the following questions
  • 19. Answers will come from specific assessments/studies: Does it work? ---- Assessment of Efficacy. How will it be delivered and how will the body react? ---- Profiling. Is it safe? --- Toxicology/safety. Is the manufacture viable and controllable? Non-clinical development activities can continue throughout the lifecycle of the product.
  • 20. Various Pharmacological Approaches to Drug Discovery: As an academic principle Pharmacology can be loosely defend as the study of effects of chemical substances on living systems. This definition is so broad that it holds all the aspects of drug discovery, ranging from details of interaction between drug molecule and its target to consequences of placing the drug in the market. Selectivity Testing: It consists of two main stages i.e. screening for selectivity and Binding assay. To determine the potency of drug, the selectivity of a compound for chosen molecular target needs to be assessed. Pharmacological Profiling: This includes the determination of pharmacodynamics effect of new compound, either on In- vitro models (which include cell lines or isolated tissues) or in- vivo models (which include normal animals, animal models of disease).
  • 21.
  • 22. The aim of Pharmacological profiling is to answer the following questions: Do the molecular and cellular effects measured in screening assays actually give rise to the predicted pharmacological effects in intact tissues and whole animals? Does the compound produce effects in intact tissues or whole animals not associated with actions on its principle molecular target? Is there a correspondence between potency of the drug at molecular level, time level and the whole animal level? # Does in-vivo duration of action match up with the pharmacokinetic properties of the drug? What happens if the drug is continuously or repeatedly given to an animal over a course of days or weeks? Does it loose its effectiveness or reveal effects not seen on acute administration and whether there is any rebound after effect when it is stopped.
  • 23. SAFETY PHARMACOLOGY: • This includes the scientific evaluation and study of potentially life threatening pharmacological effects of a potential drug which is unrelated to the desired therapeutic effect and therefore may present a hazard. • These tests are conducted at doses not too much in excess of the intended clinical dose. • Safety pharmacology seeks to identify unanticipated effects of new drugs on major organ function (i.e. secondary pharmacological effects). • It is aimed at detecting possible undesirable or dangerous effects of exposure of the drug in therapeutic doses. • The emphasis is on acute effects produced by single-dose administration rather than effects on chronic exposure as in toxicological studies.
  • 24. TOXICOLOGICAL APPROACHES TO DRUG DISCOVERY: 1. Acute Toxicity: Studies should be carried out in at least two species, usually mice and rats using the same route as intended for humans. In addition, at least two more routes should be used to ensure systemic absorption of the drug; this route may depend on the nature of the drug. Mortality should be looked for up to 72 hours after parenteral administration and up to 7 days after oral administration. The symptoms, signs and mode of death should be reported, with appropriate macroscopic and microscopic findings where necessary.
  • 25. 2. Long Term Toxicity: These studies should be carried out in at least two mammalian species and out of these two mammalian species one should be a non rodent. The duration of study will depend on the factor that whether the application is for marketing permission or for clinical trial, and in the later case, on the phases of trials. If a species is known to metabolize the drug in the same way as humans, it should be preferred in long-term toxicity studies. The drug should be administered 7 days a week by the route intended for clinical use in humans. A control group of animals, given the vehicle alone, should always be included, and three other groups should be given graded doses of the drug; the highest dose should produce observable toxicity, the lowest dose should not cause observable toxicity, but should be comparable to the intended therapeutic dose in humans or multiple of it.
  • 26. Phase 0 Pharmacodynamics and pharmacokinetics in human- phase 0 trials are optional first in human trials singles of therapeutic doses of the study drug or treatment are given to a small number of subjects ( typically 10 to 15) together preliminary data pharmacodynamics and pharmacokinetics parameters Phase I Trials Screening for safety phase one consists of usually in health valentines determine safety and dosing ( testing within a small group of people typically 20-80 ) Phase II Trials Establishing the preliminary efficacy of drug usually against a placebo - fresh to is used to get an initial grading of efficacy and father explore safety in small number of patients having the disease targeted by the NCE ( testing with large group of people typically 100- 300 ) Phase III Trials Final confirmation of safety and efficacy - phase 3 is large pivotal trials to determine safety and efficacy in sufficiently large number of patients with targeted disease. if safety and efficacy are adequately proved, clinical testing may stop at the step at the NCE advances to the new drug application ( NDA) stage ( testing with large group of people typically 1000 to 3000 ) Phase IV Trials Safety studies during sales - phase 4 is post approval trials that are sometimes a condition attached by the FDA, also called post market surveillance studies.
  • 27. Name of the Application For What IND (Investigational new drug) It is an application filled to the FDA in order to start clinical trials in humans on the basis of the data obtained from the printer clinical trials. NDA ( new drug application) If clinical studies confirm that a new drug is relatively safe and effective and will not pose a reasonable risks to patients. ANDA ( abbreviated new drug application ) It is made for approval of genetic drugs. BLA ( biologic license application ) Biology products are approved for marketing under the provisions of the public health service act.
  • 28.
  • 29.
  • 30. Investigational New Drug (IND) Application: Investigational New Drug (IND) is a program by which any Pharmaceutical company can approach to obtain permission for the initiation of human clinical trials and an experimental drug across state lines (usually to clinical investigators) before a marketing application for the drug has been approved. An investigational new drug (IND) application is to provide the data showing that it is reasonable to begin tests of a new drug on humans. The IND application is also the vehicle through which a sponsor advances to the next stage of drug development known as clinical trials. Current Federal law requires that a drug be the subject of an approved marketing application before it is transported or distributed across state lines (Clinical Investigators).
  • 31. Because a sponsor will probably want to ship the investigational drug to clinical investigators in many states, it must seek an exemption from that legal requirement. The IND application is the means through which the sponsor technically obtains this exemption from the FDA During a new drug's early preclinical development, the sponsor's primary goal is to determine if the product is reasonably safe for initial use in humans and if the compound exhibits pharmacological activity that justifies commercial development. When a product is identified as a viable candidate for further development, the sponsor then focuses on collecting the data and information necessary to establish that the product will not expose humans to unreasonable risks when used in limited, early-stage clinical studies. FDA‘s role in the development of a new drug begins when the drug's sponsor (usually the manufacturer or potential marketer), having screened the new molecule for pharmacological activity and acute toxicity potential in animals, wants to test its diagnostic or therapeutic potential in humans.
  • 32. At that point, the molecule changes in legal status under the Federal Food, Drug, and Cosmetic Act and becomes new drug subject to specific requirements of the drug regulatory system.
  • 33. Types of IND Applications: 1. Investigator IND Application, 2. Emergency use IND application, 3. Treatment IND application, 4. Screening IND application. 1.Investigation IND application: In this an application is submitted by a physician, who both initiates and conducts an investigation, and under whose immediate direction the investigational drug is administered or dispensed. A physician might submit a research IND application to propose studying of: An unapproved drug, An approved product for a new indication or An approved product in a new patient population.
  • 34. 2. Emergency use IND Application: This application allows the FDA to authorize use of an experimental drug in an emergency situation that does not allow time for submission of an IND application, in accordance with 21CFR, Sec. 312.23 or Sec. 312.20. It is also used for patients who do not meet the criteria of an existing study protocol, or if an approved study protocol does not exist. In such a case, FDA may authorize shipment of the drug for a specified use in advance of submission of an IND application. 3. Treatment IND Application: This application is submitted for experimental drugs showing promise in clinical testing for serious or immediately life-threatening conditions while the final clinical work is conducted and the FDA review takes place. A drug that is not approved for marketing may be under clinical investigation for a serious or immediately life- threatening disease condition in patients for whom no comparable or satisfactory alternative drug or other therapy is available. In the case of a serious disease, a drug ordinarily may be made available for treatment use during phase III investigations or after all clinical trials have been completed. In the case of an immediately life-threatening disease a drug may be made available for treatment use earlier than phase III but ordinarily not earlier than phase II.
  • 35. 4. Screening IND Application: It is flied for multiple, closely related compounds in order to screen for the preferred compounds or formulations. The preferred compound can be developed under a separate IND. It can also be used for screening different salts, esters and other drug derivatives that are chemically different, but pharmacodynamically similar. IND Review and Report: During this time, FDA has an opportunity to review the IND application for safety to assure that research subjects will not be subjected for unreasonable risk. The report evaluates on the various factors like Medical Review, Chemistry review, Pharmacology/Toxicology review, Statistical analysis and Safety review.
  • 36. Layout chart for IND Application
  • 37. Investigators Brochure (IB):- The Investigator‘s Brochure (IB) is a compilation of the clinical and non-clinical data on the investigational product(s) that are relevant to the study of the product(s) in human subject. 1.Purpose of Investigator‘s Brochure (IB) : Its purpose is to provide information to the investigators and others involved in the trial such as the dose, dose frequency/interval, methods of administration and safety monitoring procedures. The IB also provides insight to support the clinical management of the study Subjects during the course of the clinical trial. The information should be presented in a concise and simple manner. IB enables a clinician, or potential investigator, to understand it and make his/her own unbiased ( without any partiality) risk-benefit assessment of the appropriateness of the promised trial. For this reason a medically qualified person should generally participate in the editing of an IB.
  • 38. 2. Contents of Investigators Brochure : 1.Table of contents. 2. Summary not exceeding 2 pages, highlighting the significant physical, chemical, Pharmaceutical, Pharmacological, toxicological, pharmacokinetic, metabolic, and clinical information available of IP. 3. Introduction: Chemical name, active ingredients, pharmacological class, anticipated therapeutic/diagnostic indication(s). General approach to be followed in evaluating the IP. 4. Description as per IP: Physical, chemical and pharmaceutical properties as per IP storage and handling as per IP. Any structural similarity with the other known compound given. 5. Non-Clinical Studies: The results of all relevant non- clinical pharmacology, toxicology, pharmacokinetic, and investigational product metabolism studies should be provided in summary form. The information provided may include: Species tested, Unit dose (e.g., milligram/kilogram (mg/kg), Dose interval, Route of administration and Duration of dosing.
  • 39. 5.1 Non-Clinical Pharmacology: A summary of the pharmacological aspects of the investigational product studied in animals should be included. 5.2 Pharmacokinetics and Product Metabolism in Animals: A summary of the pharmacokinetics (ADME) and biological transformation and disposition (getting a drug into its appropriate position in the body and in an appropriate concentration) of the investigational product in all species studied should be give 5.3 Toxicology: (The study of the adverse effects of chemicals on animals): A summary of the toxicological effects found in relevant studies conducted in different animal species. (Single dose, Repeated dose, Carcinogenicity, Special studies (irritancy, sensitization), Reproductive toxicity).
  • 40. 6. Effects in Humans: A thorough discussion of the known effects of the investigational product(s) in humans should be provided, including information on Pharmacokinetics, metabolism, Pharmacodynamics, dose response, safety, efficacy, and other pharmacological activities like Pharmacokinetics and Product Metabolism in Humans 7. Summary of Data and Guidance for the investigator: This section should contain non-clinical and clinical data of IP. Investigators Brochure (IB) provides the Investigator a clear understanding of the possible risks, adverse reactions, observations and precautions needed for the clinical trial.
  • 41. NEW DRUG APPLICATION (NDA) Definition: An NDA is an application to permit the sale and marketing of a new drug. The vehicle through which drug sponsors formally propose that the regulatory body approves a new pharmaceutical products for sale and marketing, and the data gathered during the animal studies and human clinical trials of an investigational new product becomes a part of the NDA. Aim of NDA: The aims of NDA include providing adequate information to permit FDA reviewers to establish the following:  Safety and effectiveness of drug,  Benefits overweigh risks,  Is the drug's proposed labeling (package insert) appropriate, and what should it contain?  Are the methods used in manufacturing (Good Manufacturing Practice, GMP) the drug and the controls used to maintain the drug's quality adequate to preserve the drug's identity, strength, quality, and purity? Risk Benefit.
  • 43. NDA Review Process: Once the application is submitted, the FDA has 60 days to conduct a preliminary review which will assess whether the NDA is "sufficiently complete to permit a substantive review". If everything is found to be acceptable, the FDA will decide if the NDA will get a standard or accelerated review and communicate the acceptance of the application and their review choice in another communication known as the 74- day letter
  • 45. Bio Equivalence Studies: BE studies are very essential to ensure uniformity in standards of quality, efficacy and safety of pharmaceutical products so that reasonable assurance can be provide for the various products containing same active ingredient, marketed by different licensees are clinically equivalent and interchangeable. Both Bioavailability and Bioequivalence focus on release of drug substance from its dosage form and subsequent absorption in circulation. Similar approaches to measure bioavailability should be followed in demonstrating bioequivalence.
  • 47. Bioavailability: Bioavailability means 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. Equivalence: It is a relative term that compares drug products with respect to a specific characteristic or function or to a defined set of standards.
  • 48. Clinical Research Protocols: It is a complete written description of and scientific rationale for a research activity involving human subjects. Sufficient information is to be gathered on the quality of the non- clinical safety to conduct the protocol and health authority/ethics committee approval is granted in the country where approval of the drug or device is sought (i.e. in past). The clinical trial design and objectives are written into a document called a clinical trial protocol. It is a document that states the background, objectives, rationale, design, methodology (including the methods for dealing with AEs i.e. Adverse Drug Events, withdrawals etc.) and statistical considerations of the study. It also states the conditions under which the study shall be performed and Look for better ways to prevent disease in people who never had the disease or to prevent a disease from returning.
  • 49. The protocols means: To clarify the research question. To compile existing knowledge. To formulate a hypothesis and objectives. To decide about a study design. To clarify ethical considerations. To apply for funding. To have a guideline and tool for the research team.
  • 50. Parts of the Protocol: 1.Title Page. 2. Signature Page. 3. Content Page. 4. List of Abbreviations. 5. Introduction/Abstract. 6. Objectives. 7. Background/Rationale. 8. Eligibility Criteria . 9. Study Design/Methods (Including Drug/Device Info). 10. Safety/Adverse Events. 11. Regulatory Guidance. 12. Statistical Section (Including Analysis and Monitoring). 13. Human Subjects Protection/Informed Consent.
  • 51.
  • 52. Biostatistics in Pharmaceutical Product Development: Biostatistics is a science where professionals collect, analyze and interpret conclusions for data. In simpler words, when we apply statistics in biological science it is called biostatistics. It requires an understanding of statistical theory and application to work out issues within biological and health science organizations. Biostatisticians typically address healthcare topics, either in the private or public sectors, and work in an office environment. The role of the biostatistician in pharmaceutical product development is more important than ever, and hence statistics professionals need to develop better approaches to solve some old limitations and some new problems
  • 53. Pharmaceutical biostatisticians analyze genetic data and disease rates. They use this information to design clinical trials to evaluate new drugs. They study populations exposed to harmful chemicals to determine their overall effects. For example, during a clinical trial, there are many phases for drug or device development. To check the outcome of the study as trial is going good or bad. biostatisticians applies concepts of statistics and do the analysis. Based on the analysis results, they draw inference about the study. So biostatistics is a method that comprises of a collection of data, summarizing, analyzing and drawing inferences.
  • 54. Basic Concepts of Biostatistics: 1.Statistical Levels and Decision Making: Hypothesis testing is a method used to determine whether the results are statistically significant or not. It is the process of drawing inferences to make decision about the sample with regards to the population as a whole. Bio statistical analyses in human clinical trials are required during the last stages of the pharmaceutical product development process. It has effectively become a hurdle for every new drug, device, and biological while on the way to market.
  • 55. 2. Sample Size: Sampling is a method used in statistical analysis of a data where a specific number of samples are taken from a population for a study. For example, health ministry wants to know the opinion or suggestions of recently implemented health program in particular region. Here the population is that region and sample is cities in that region. In the demonstration of efficacy of a new product, achieving p-value ≤ 0.05 is critical while comparing the new product to the control group. It is observed that most products with modest potential therapeutic benefit complies this specification only if the sponsor of the product invest enough money and/or time to conduct a very large trial. Sample size can overcome the limitations of such benefits and hence is the true decision criteria for effectiveness. Use of huge financial investment is not the best model for the evaluation and approval of new products. Product approval should not be based on the financial strength of the sponsor and/or product's future revenue potential. Rather, safety concerns of clinical trial participants and the future subjects of the product are the important elements. Thus, sound statistical criteria should be used to quantify sample size and to ensure public health concerns
  • 56. 3 Data Collection: it is first thing in any kind of analysis. Data are the actual pieces of information that we collect through experiments. It can be categorized into categorical and numerical data form. Categorical data are defined in categories or groups. For example, male or female, yes or no, etc. Numerical data are classified into discrete and continuous data. Discrete data are the data which take certain values. For example, number of patients, number of devices etc. Continuous data are the data which can take any values either fractional or decimal. For example, blood pressure level, height and weight of the patient etc. Data collection in a clinical trial begins as clinical information is collected from subjects. Initially it reflects some component of truth about treatment effects, and some component of variability or error. Statistical analysis techniques can estimate the magnitude of the error component. This must be used as a metric to estimate the size and reliability of the truth component.
  • 57. Biostatisticians generally require consistent and relatively simple clinical assessments to create data tabulations and perform analyses. As they are engaged in data cleaning raw data often does not meet this expectation and thus resulting altered data consists of truth, error and systematic bias. The impact of systematic bias introduced during the cleaning process is not normally assessed statistically. 4. Measurement System Analysis: Measurement system analysis helps to detect the amount of variation existing within a measurement system. Suppose during the initial clinical trial phase of a particular product, if the data are not recorded properly, and if further analysis is to be carried out then consequences of it would be a failure at final phases. The intention of clinical trials is to accurately predict benefits and risks of new drug product to the broad population of subjects, while protecting the safety of those subjects who volunteer to provide the data needed to make that prediction.
  • 58. Experimental Design in Clinical Trials: As seen under clinical trials section, these trials for a new drug, device, or biological typically are conducted from Phase-l to Phase-3 prior to NDA submission to a regulatory agency for approval. Each phase involves increase in number of subjects sequentially. Biostatistician is involved in the design and analysis of these studies. Biostatistician in collaboration with medical, regulatory and data management experts defines what to measure, how often to measure it, how to select subjects and randomly assign treatments to them, and how to analyze the outcome. All these things are pre-specified in the clinical protocol, including the expected outcome, all analysis strategies, and the rationale for the number of subjects to be incorporated in the studies.
  • 59. 1.Sample Size: In order to establish the sample size, a statistician is consulted at the beginning of a trial. Deciding required sample size is critical for Phase-3 trials. This is because statistical significance in the final analyses at this phase is required to support a submission for MAA for approval. Insufficient sample size may lead to failures for drugs to appear as adequately safe and effective for approval. The outcome of Phase-2 trial provides enough evidence to establish the required sample size for Phase-3 trials. The overall results from sample reflect both overall population response and some degree of random variability. The larger the sample size, the smaller the effect of the variability on the overall results. Inferential statistics helps to quantify uncertainty in effect of the variability. The statistical power measures the degree to which the proposed sample size can overcome unpredictable variability in subject response, as well as the degree of confidence that the results of the sample will approximate the future response of the population.
  • 60. 2. Sensitivity and Cost: The increase in the sample size in a clinical trials increases the level of assurance that the drug is an effective drug and ultimately the statistical significance when the results are analyzed using statistical methods. As sponsor has some financial and time constraint, sample sizes are always a compromise between the ideal and the affordable. This is crucial stage where a biostatistician is involved to define the parameters for sample size. The sensitivity of a trial to detect a statistically significant value also depends on sample size. Since all other things are almost equal, the larger the sample size, the less effective a drug must be to show a significant, and potentially approvable, result. Financial investment in clinical research is an important factor for obtaining marketing approval.
  • 61. 3. Random Sampling: The random sampling in clinical trials has two major requirements for inferential statistics to forecast population benefits. First, it is necessary that the subjects in clinical trials must be randomly selected and it must be a representative sample of the population of future subjects in the total population. Second, it is essential that for comparative trials, treatments must be randomly assigned to subjects in clinical trial. Commonly major trials follows later requirement, and in some trials the earlier requirement is complied.
  • 62. 4. Data Refinement: The CRFs are developed to increase clinical database to fulfill the requirements of the statistician for analysis and reporting and the sponsor to establish sufficient evidence for approval. The recommendations to avoid bias in data refinement and categorization activities are given below: (i) Clinical trial data captured during study should be categorized into its objectivity or subjectivity, immediacy or delay of collection and source. (ii) Apply objective data cleaning criteria, for example, range checks and consistency comparisons. (iii) For subjective data exercise extreme caution when questioning values (especially when a time has elapsed since its collection). (iv) Formalized coding schemes should be relevant to the drug under study. (v) Data cleaning efforts for variables that are not Identified as critical should be minimized.
  • 63. DATA PRESENTATION FOR FDA SUBMISSIONS: Study data standards describe a standard way to exchange clinical and non-clinical study data. These standards provide a consistent general framework for organizing study data, including templates for datasets, standard names for variables; identify appropriate controlled terminology and standard ways of doing calculations with common variables. Data standards also help FDA receive, process, review, and archive submissions more efficiently and effectively. FDA has been working towards a standardized approach to capture, receive and analyze study data. Standardization of study data is vital to integrate pre- marketing study data and post-marketing safety data to improve public health and patient safety. Central to this vision is the creation of an enterprise data infrastructure (Janus) within FDA to improve the management of all structured scientific data.
  • 64. Data Standards: Data standards can be divided into two categories: 1. Exchange Standards: Exchange standards provide a consistent way to exchange information. Exchange standards help to ensure that the sending and the receiving systems both understand unambiguously what information is being exchanged. 2. Terminology Standards: Terminology standards provide a consistent way to describe concepts. For example, the Unique Ingredient Identifiers (UNII), developed by the FDA, provides a consistent way to describe substances in foods and drugs. Vocabulary developed National Institute of Cancer describes terminology related to cancer.
  • 65. MANAGEMENT OF CLINICAL STUDIES: Clinical trial management is most simply defined as the process that an organization follows to ensure that quality (defined as minimized risks and clean data) is delivered efficiently and punctually. It refers to a standards-driven process that a project manager initiates and follows in order to successfully manage clinical trial sites, clinical research associates, and workflow by using clinical trial management tools or software prolonged timelines and heavy costs related to large trials have been prompted a new focus on more eminent clinical trial management. It is possible to dramatically reduce the total cost of a clinical trial by 60% 90% without compromising the scientific validity of the results.
  • 66. Life Cycle of Clinical Trial Project: A more accurate control, regardless of the therapeutic area or trial stages (all the way through from 'preclinical’ phase 1 to 'post- approval' phase 4 studies), is ensured by typically breaking down the life cycle of each clinical trial project into 4 phases: Conceptual, Planning, Implementation and Analysis. Clinical Trial Protocol: A protocol is a document that describes the purpose, design, methodology, statistical considerations and organization of a study, and provides basic information and rationale for the clinical study. The contents that should be present in the protocol are described by the GCP. The protocol writing is a task for one person, usually the principal investigator, not a committee. There are various challenges of Project management in clinical trials. Clinical trials all need the same coordinated processes and systems, irrespective of the size, scope, costs, or period. The key challenge is then to implement and maintain effective management systems and techniques in response to the needs of the trial project.