SlideShare une entreprise Scribd logo
1  sur  42
Télécharger pour lire hors ligne
UNIT 5: Documetation 
Documentation is a process that involves the systematic interaction of people, events and documents to 
create the records of the organization / corporation. 
It is a collection of document (a piece of written or printed matter) relating to a piece (or) events. 
Documentation is an important aspect in pharmaceutical industry irrespective of the fact the manufacturing 
plant is involved in production of dosage form (or) active ingredient. 
Manufacturer of a drug, ultimately needs quality of his product (or) finished goods. Therefore a good 
documentation is essential part of a quality control system, hence every manufacturer of drugs should have 
good documentation. 
Its aim is to define the specifications for all materials, method of manufacture and control, to ensure that all 
personnel concerned with manufacture know the information necessary to decide whether or not to release 
a batch or a drug for sale and-to provide an 
audit trail that shall permit investigation of the history of any suspected defective batch. 
DOCUMENT: 
As discrete packages of recorded information are the tools of quality assurance. 
Used to establish standard specification and processes that assures compliance with standard specifications 
and processes. 
Quality cannot be assured in a regulated industry without good documents and good documentation 
practices. 
A document system is an interdependent, interrelated set of documents, each with a defined purpose and a 
consistent format. 
OBJECTIVES OF DOCUMENTATION: 
· To define all materials specifications. 
· To define manufacture and control methods. 
· To ensure all personal of manufacture and control department know what to do, and when to 
do. 
· To ensure that personal authorized to release a batch for sale (or) reject, it have all 
information to take decision. 
· To provide information during investigation, if a batch is suspected to have 
defect(s). 
ESSENTIAL CHARACTERISTICS: 
To design an effective & efficient documentation process first identify the important characteristics of 
processing event. 
· The document directing the manufacturing event fulfills current regulatory commitments to 
the agency; it is appropriately written, reviewed and approved. 
· The directive document is appropriate for the task to be performed. 
· The data is authentic; the individual responsible for performing the work has entered the 
data on an appropriate data collection document. 
· The data is accurate. 
· The data is complete. There is no missing information, and there is no work as yet 
uncompleted that will impact the occurrence of the data presented. 
· The data is legible, consistently recorded and trustworthy. 
· The data collected fulfills expectations (specifications). 
· The data is accessible to those who need to review it, audit it or use it to perform trending 
analysis. 
· The original data and the original documents (manufacturing record) is retrievable for 
review or audit.
· The original manufacturing record is secure. 
Proper control of documentation also requires that responsibility for each processing step to be 
assigned. 
“All documentation should be legible, clean, readily identifiable, retrievable and maintained in 
facilities that provide a suitable environment to minimize deterioration or damage and to prevent 
loss”. 
WHY DOCUMENTATION: 
· There is a saying in the pharmaceutical industry: 'if it hasn't been documented, then it hasn't 
happened!' 
· Good documentation practice constitutes an essential part of the QA system. 
· Documentation system must be proactive vehicle of communication. 
“YOUR DOCUMENTATION IS AN ADVERTISEMENT FOR YOUR WORKˮ. 
WHERE DOCUMENTATION ? 
· Documentation during Project Design. 
· Documentation during Construction Phase. 
· Documentation during Commissioning and start-up. 
· Documentation during Qualification and Validations. 
· Documentation during Commercial Production. 
· Documentation during Testing and Release. 
· Documentation for Regulatory submissions. 
TYPES OF DOCUMENTS: 
There are three types of Documents: 
· Commitment documents. 
· Directive documents. 
· Record documents. 
· Commitment Documents : Relationship between industry and the regulatory authorities. 
EX: New Drug Applications (NDAs), Drug Master Files (DMFs) etc. 
· Directive Documents : Relationship between the Management and Employees. 
EX: Specifications, STPs, Standard Operating Procedures (SOPs), Medicinal Product 
Records (MPRs) etc. 
· Record Documents : Relationship between the Employees and the Work they perform. 
EX: Protocols, Batch Production Records (BPRs), Log Books, Calibration Records etc. 
· Commitment Documents: 
· These presents corporate goals, expectations and standard of practice. 
· It describes what to do. 
· Consensus of purpose, direction and authorization for projects. 
· It organize the work in a manner that assures efficient and effective work flow. 
· Commitments documents can also be written to establish internal commitments. 
· These documents such as master plans; organize and prioritize the work in a manner that 
supports good business practice as well as regulatory compliance. 
· Written to lead and guide the work and workers. 
· To be used as active documents that are consulted and followed routinely.
· Content must be consistent and rigorous in order to lead and guide the work and the 
workers. 
· Must describe the work as it will be done. 
· Deviations from regulatory submissions can have signification impact on product quality. 
· Any deviation must be documented and managed. 
· New Drug Applications (NDAs): 
The New Drug Application (NDA) is the vehicle in the United States through which drug 
sponsors formally propose that the FDA approve a new pharmaceutical for sale and marketing. The goals 
of the NDA are to provide enough information to permit FDA reviewers to establish the following: 
· Is the drug safe and effective in its proposed use(s) when used as directed, and do the 
benefits of the drug outweigh the 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? 
· Drug Master Files (DMFs): 
Drug Master File or DMF is a document prepared by a pharmaceutical manufacturer and submitted solely 
at its discretion to the appropriate regulatory authority in the intended drug market. There is no regulatory 
requirement to file a DMF. However, the document provides the regulatory authority with confidential, 
detailed information about facilities, processes, or articles used in the manufacturing, processing, 
packaging, and storing of one or more human drugs. 
Drug Master File (DMF) is a document containing complete information on an Active Pharmaceutical 
Ingredient (API) or finished drug dosage form. It is known as European Drug Master File (EDMF) or 
Active Substance Master File (ASMF) and US-Drug Master file (US-DMF) in Europe and United States 
respectively. 
The DMF contains factual and complete information on a drug product's chemistry, manufacture, stability, 
purity, impurity profile, packaging, and the cGMP status of any human drug product. 
· Directive Document: 
· Working documents that establish the standards for resources, processing, products & 
quality system. 
· Describe how to do it……??? 
· Describe how to do routine work. 
· Several types of directive documents. 
· The different types are determined by the specific, functional purpose of the document in the 
document system. 
· In order to facilitate the development, production, testing and distribution of a product in a 
defined manner. 
· Reviewed and approved by both management and the individuals responsible for performing 
the work. 
· Specifications: 
A document specification contains several parts: a description of the audience(s) for the document, a 
detailed outline giving the structure and contents of the document, and a work plan showing who is 
responsible for each part of the document and what the deadlines are for completing each task. For large 
documents, there may other managerial information, such as number of pages allocated for each section, 
graphics budgets, printing costs, and so forth. We do not expect you to produce this level of detail in your 
document specification. 
There are three purposes for document specifications:
In the workplace, formal document specifications serve three important functions: economy of effort, work 
planning, and writing organization. 
· Economy of effort: 
First, document specifications are used to help you reduce the amount you have to write. When you are 
requested to write a major report, quite often you will be told something like, ‟Do a draft and then come see 
me with it”. After talking with your boss, you will have to revise the draft completely so that it will have in 
it what the boss really wants, since he/she hadn't thought about the project carefully until reading your 
draft. You can often save yourself (and your boss) much work if you write a detailed document 
specification instead. A document specification is much easier to create, change, revise, and add to than a 
draft is. 
· Work planning: 
The second major function of a document specification is work planning. This can mean either budgeting 
your own time, or, in large formal reports, distributing the work among many people. Multi-author writing 
is probably the most common form of workplace writing. The more you know about the specification, the 
more likely you are to get only your fair share of the writing and no more. For the purposes of the class, the 
document specification will serve to distribute the work between you and your partner. You should bear 
three things in mind in dividing the work: even work load, respective areas of expertise, and getting things 
done in the order that you need them. These points sound obvious, but achieving a fair distribution of the 
work is not always easy. It involves keeping to a schedule, for one thing, and staying in close touch with 
your partner, for another. 
· Writing Organization: 
The third major function is the organization of the report itself. 
· Standard Operating Procedures (SOPs): 
Standard Operating Procedures (SOPs) are issued to specifically instruct employees in areas of 
responsibility, work instructions, appropriate specifications and required records 
A Standard Operating Procedure (SOP) is a set of written instructions that document a routine or repetitive 
activity followed by an organization. The development and use of SOPs are an integral part of a successful 
quality system as it provides individuals with the information to perform a job properly, and facilitates 
consistency in the quality and integrity of a product or end-result. The term “SOP” may not always be 
appropriate and terms such as protocols, instructions, worksheets, and laboratory operating procedures may 
also be used. For this document “SOP” will be used. 
SOPs describe both technical and fundamental programmatic operational elements of an organization that 
would be managed under a work plan or a Quality Assurance (QA) Project Plan [EPA Requirements for 
QA Project Plans (QA/R-5) (EPA 2001a)], or Chapter 5 of the EPA Quality Manual for Environmental 
Programs, (EPA Manual 5360 A) and under an organization’s Quality Management Plan [EPA 
Requirements for Quality Management Plans (QA/R-2) (EPA 2001b)], or Chapter 3 of the EPA Quality 
Manual. This document is designed to provide guidance in the preparation and use of an SOP within a 
quality system. 
Purpose: 
SOPs detail the regularly recurring work processes that are to be conducted or followed within an 
organization. They document the way activities are to be performed to facilitate consistent conformance to 
technical and quality system requirements and to support data quality. They may describe, for example, 
fundamental programmatic actions and technical actions such as analytical processes, and processes for 
maintaining, calibrating, and using equipment. SOPs are intended to be specific to the organization or 
facility whose activities are described and assist that organization to maintain their quality control and 
quality assurance processes and ensure compliance with governmental regulations. 
If not written correctly, SOPs are of limited value. In addition, the best written SOPs will fail if they are not 
followed. Therefore, the use of SOPs needs to be reviewed and re-enforced by management, preferably the 
direct supervisor. Current copies of the SOPs also need to be readily accessible for reference in the work
areas of those individuals actually performing the activity, either in hard copy or electronic format, 
otherwise SOPs serve little purpose. 
· Record document: 
Protocol: 
Protocols are written records clearly defining the objectives and methods that will be used for the validation 
programs. An important part of the protocol is the description of the testing method including who will test 
the system, how they will test it and what data is to be collected and reported. 
Computerized system protocols often include the three distinct stages as described in PMA reports: 
Installation Qualification (IQ), Operational Qualification (OQ),and Performance Qualification (PQ). 
Protocol Changes are documented requirements specifying who and how changes to parameters, 
thresholds, and acceptance criteria are made after approval. It is not impossible to make changes after or 
during testing, but these changes must be properly implemented and approved to be validatable. 
What Good Documentation requires: 
APPROVAL - this applies particularly to work instructions, procedures, manufacturing formulae and 
specifications. 
Approval should be by the relevant technical, management and quality personnel, to ensure that documents 
comply with the principles of GMP and the specific product marketing and manufacturing authorizations. 
CLARITY - they should not be open to misinterpretation by the users. They should be written in a way that 
makes them easy to check, particularly when they will form part of a product manufacturing history. 
Good documentation design will help to minimize errors. 
REGULAR REVIEW AND UPDATE - documents must be kept up-to-date with changes in regulations or 
processes and should be distributed in a controlled manner to ensure that only the most recent versions are 
available for use. They must also be available to those who need them, where they need them! 
FORMAL PRESENTATION - controlled documents should be prepared in accordance with a written 
procedure, now a days probably using a computerised documentation control system. 
Records should be made at the time of each action - do not rely on memory for their completion. 
Records relating to manufacturing or testing operations should be kept for at least one year after their 
expiry. 
If documents or data are stored electronically, the computer system must be validated to assure data 
security and integrity. 
Provisions must also be made to retrieve the stored data, possibly years after they have been generated. 
PROTOCOL FOR DOCUMENTATION: 
Protocol is a detailed plan and instructions to the employees working in pharmaceutical industry. 
It is a guidebook for those involved in work. 
The manufacturing records relating to manufacture of sterile products shall indicate the following details:- 
(1) Serial number of the Batch Manufacturing Record. 
(2) Name of the product. 
(3) Reference to Master Formula Record. 
(4) Batch/Lot number. 
(5) Batch/Lot size. 
(6) Date of commencement of manufacture and date of completion of manufacture. 
(7) Date of manufacture and assigned date of expiry. 
(8) Date of each step in manufacturing. 
(9) Names of all ingredients with the grade given by the quality control department. 
(10) Quality of all ingredients. 
(11) Control reference numbers for all ingredients. 
(12) Time and duration of blending, mixing, etc. whenever applicable. 
(13) pH of solution whenever applicable. 
(14) Filter integrity testing records. 
(15) Temperature and humidity records whenever applicable.
(16) Records of plate-counts whenever applicable. 
(17) Results of pyrogen and/or bacterial endotoxin & toxicity. 
(18) Results of weight or volume of drug filled in containers. 
(19) Bulk sterility in case of aseptically filled products. 
(20) Leak test records. 
(21) Inspection records. 
(22) Sterilization records including autoclave leakage test records, load details, date, duration, temperature, 
pressure, etc. 
(23) Container washing records. 
(24) Total number of containers filled. 
(25) Total numbers of containers rejected at each stage. 
(26) Theoretical yield, permissible yield, actual yield and variation thereof. 
(27) Clarification for variation in yield beyond permissible yield. 
(28) Reference numbers of relevant analytical reports. 
(29) Details of reprocessing, if any. 
(30) Name of all operators carrying out different activities. 
(31) Environmental monitoring records. 
(32) Specimens of printed packaging materials. 
(33) Records of destruction of rejected containers printed packaging and testing. 
(34) Signature of competent technical staff responsible for manufacture and testing. 
In addition to the routine good manufacturing practices documentation, manufacturing records shall show 
the following additional information:- 
(1) Temperature and humidity in the manufacturing area. 
(2) Periodic filled weights of the formulation. 
(3) Records of rejections during on line check weighing. 
(4) Records of rejection during spray testing. 
REFERENCES: 
· www.s uperiorcontrols.com/news.php?id=31 & page=4 
· http://www.medicalnewstoday.com/articles/172522.php 
· http://www.drugs.com/nda/acetavance_090715.html 
· http://www.sciencemag.org/cgi/content/full/326/5951/370 
· http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/u 
cm122886.htm 
Formulation development for global fillings 
THE IMPACT OF WORLD REGULATIONS TO DEVELOP AND LAUNCH HARMONIZED 
COSMETIC PRODUCTS 
Business Strategy 
· Since long time ago, we have been challenged: 
“ How can we develop a cosmetic product that can have its inventory ready to be exchanged around 
the globe.” 
· Same package with all information in 
· Same formula 
“In other words: How to make one fit all” 
Understand what is required in each country to sell cosmetics 
Global Cosmetic Regulation Scenario 
We have a lack of global harmonization on Cosmetics: 
· USA
· Europe 
· Asia 
· Latin America 
Robust R&D system is required 
Latin America 
· Mercosur 
· CAN 
· Central America 
· Mexico 
· Chile 
· Panama 
· All others 
Mercosur 
· Argentina, Brazil, Paraguay and Uruguay 
· PreMarketing approval process 
· Lists of ingredients harmonized 
· In place process to update the list 
· Unique requirements still present in each country 
· Administrative procedures to get product registration are not harmonized 
· Requirements change country by country 
· Labeling requirements not harmonized 
· Claims /Safety support data requirements not harmonized 
CAN 
· Bolivia, Colombia, Ecuador, Peru and Venezuela* 
· PreMarketing Approval process 
· Recognition Process in place 
· Adopts List of Ingredients from Europe and USA 
· Administrative procedures to get product registration are almost quite harmonized: 
· Requirements quite clear 
· Labeling requirements almost quite harmonized 
· Claims /Safety support data requirements almost quite harmonized 
Central America (RTCA) 
· Costa Rica, El Salvador, Guatemala, Honduras, Nicaragua. 
· New Harmonized cosmetic regulation since early 2009 
· PreMarketing approval process 
· Adopts List of Ingredients from Europe and USA 
· Administrative procedures to get product registration are not harmonized: 
· Requirements change country by country 
· Labeling not harmonized 
· Claims /Safety support data requirements not harmonized 
Mexico 
· Pos-Marketing Surveillance process for cosmetics 
· Pre-Marketing approval process for Hygienic
· Has list of allowed, restricted and not allowed ingredients updated on March 21, 2007 
· Specific labeling requirements ( INCI translated to Spanish) 
Chile and Panama 
· PreMarketing approval process 
· Adopts List of Ingredients from Europe and USA 
· Specific labeling requirements 
R&D Process 
· Product Development 
· R&D systems should be able to provide to the Formulator the ingredient’s regulation 
around the global/region so that only approved ones can be used 
· Product Safety: 
· Make sure that ingredients that are not regulated are safe. 
· Make sure that final formulation is safe 
· Make sure claims are substantiated 
Regulatory Affairs 
· Make sure to update database system 
· Ingredients update 
· Safety test requirements 
· Specific country/region product registration requirements: 
· Words/expression restriction / forbidden 
· Claims requirements 
· Unique requirements
Documentation
Documentation
Harmonized Product 
· Last but not least 
· Define Manufacturing strategy 
· This will drive the last regulatory requirements to have harmonized product: 
· Same package 
· Same formula 
· Same label copy 
· Approved in the countries according strategy 
The impact of world regulation to develop and launch harmonized cosmetic products 
· Time to launch harmonized product is too long impacting: 
· Marketing strategy 
· Supply Chain strategy 
· Adding Premium costs 
· Service issues 
Patents are exclusive property rights in intangible creations of the human mind. They exist only as provided 
in the laws of sovereign states, and can be enforced only to the extent that application has been made and a 
patent granted covering the territory of an individual state. Patent rights are limited in duration, with the 
global standard being 20 years from the date of application. The new product, article of manufacture or 
process described in the patent application must be something that has never been previously disclosed 
anywhere in the world and something that would not be obvious to a person ordinarily skilled in the field 
involved. Determinations of whether these requirements have been met are made by comparing the claims
of the patent applicant against the body of published literature in the field, including previously issued 
patents. This process is called examination, and it assures that no one is able to claim patent rights on 
anything that already is existence. 
Patents work differently indifferent industries. In the electronic industry patents are often shared among 
competitors through pooling or cross licensing. This sharing is necessary because a given product often 
contains many patented technologies. However, in the pharmaceutical, chemical and biotechnology 
industries the patent normally equals the product, and protects the extensive investment in research and 
clinical testing required before placing it on the market. Patent protection for chemical and pharmaceutical 
products is especially important compared with other industries because the actual manufacturing process is 
often easy to replicate and can be copied with a fraction of the investment of that required for the research 
and clinical testing. 
The extensive cost required to produce a new pharmaceutical product has meant that private sector 
investment in pharmaceutical innovation has been disproportionately directed to products meeting the 
needs of patients in developed countries, particularly in the United States, which combines strong patent 
protection with a market free of price controls. 
Until the TRIPS Agreement in 1994 many developing countries provided no patent protection for 
pharmaceutical products. And, while countries that have joined the WTO have obligated themselves to 
provide such protection, least developed countries are not required to meet this obligation until 2016. The 
continuing lack of patent protection for pharmaceutical products makes it very difficult to establish 
research-based industries in most developing countries. Most medical research in these countries takes 
place in the public sector. The lack of any means of patenting these inventions and the related lack of 
experience in licensing them to the private sector, suppresses the development of commercial enterprises 
focused on alleviating the disease burdens common to developing countries. 
The controversy over availability of patented therapies for the treatment of HIV disease has resulted 
renewed interest in the compulsory licensing of pharmaceutical products. After two years of discussion, the 
WTO Council recently affirmed that the TRIPS Agreement permits such compulsory licenses in health 
emergencies, even in cases where the compulsory license is for an imported product. However, to date, no 
compulsory licenses actually have been issued, even though the threat of compulsory licensing has been 
used as a means of seeking lower prices. 
One danger in compulsory licensing is that it will discourage further the commercial R & D necessary to 
new drugs to fight global epidemics. Another danger is that compulsory licensing can be used to seek price 
levels below what a given national market is capable of supporting, further concentrating the burden of 
financing pharmaceutical innovation on developed country consumers and discouraging development of 
drugs targeted at the disease burdens of countries using compulsory licenses. 
There are promising developments in countries such as India and Brazil that are beginning to use patents to 
develop commercial pharmaceutical industries that produce products directed at local diseases and 
available at price that patients in those countries can afford. Foundations and nonprofit organizations such 
as the Bill and Melinda Gates Foundation and One World Health, Inc. are supporting such efforts. These 
efforts show that developing countries have the capacity to build research-intensive pharmaceutical 
industries capable of operating profitably in the conditions of the local market. However, for such local 
industries to take root and grow, effective patent protection must be made available, the commercialization 
of publicly funded research must be encouraged, and compulsory licensing must be kept to a minimum. 
Wealthy countries can assist this process by subsidizing local markets for the purchase of drugs through the 
Global Fund, and by direct programs of assistance such as that recently proposed by President Bush. 
Consumers in all countries can share the burden of drug development equitably by paying for medicine at a 
price level consistent with their means, rather than attempting to shift the costs of drug development to 
others. 
REFERENCES: 
1. http://www.personalcarecouncil.org
2. http://ec.europa.eu/enterprise/cosmetics/ 
3. http://www.cirsafety . 
org/ 
NDA 
INTRODUCTION: 
New Drug Application is a Critical component for drug approval process which required to submit to 
USFDA before drug commercialization. The data gathered during the animal studies and human clinical 
trials of an Investigational New Drug (IND) become part of the NDA. The regulation and control of new 
drugs in the United States has been based on the New Drug Application (NDA). Since 1938, every new 
drug has been the subject of an approved NDA before U.S. commercialization. 
Goals of the NDA: 
To provide enough information to permit FDA reviewer to reach the following key decisions: 
· Whether the drug is safe and effective in its proposed use(s), and whether the benefits of the 
drug outweigh the risks. 
· Whether the drugs proposed labeling (package insert) is appropriate, and what it should 
contain. 
· Whether the methods used in manufacturing the drug and the controls used to maintain the 
drug's quality are adequate to preserve the drug's identity, strength, quality, and purity. 
NDA Classifications: 
· New Molecular Entity 
· New Salt of Previously Approved Drug (not a new molecular entity) 
· New Formulation of Previously Approved Drug (not a new salt OR a new molecular entity) 
· New Combination of Two or More Drugs Already Marketed Drug Product - Duplication 
(i.e., new manufacturer) 
· New Indication (claim) for Already Marketed Drug (includes switch in marketing status 
from prescription to OTC) 
· Already Marketed Drug Product - No Previously Approved NDA. 
Assembling Applications for Submission: 
Assembling Applications for Submission the FDA requires drug sponsors to submit multiple copies of the 
NDA The archival copy, the review copy, the field copy. In 1997 the FDA Center for Drug Evaluation and 
Research (CDER) published guidelines that allow sponsors to submit NDAs electronically instead of on 
paper. 
The Archival Copy: 
The Archival Copy Contains all sections of the NDA, including the cover letter, Form FDA-356h 
(Application to Market a New Drug, Biologic, or an Antibiotic for Human Use), the administrative 
sections, Comprehensive NDA index, and All technical sections. It must contain four copies of the 
Labeling section. It must contain three additional copies of the CMC and Methods Validation Package in a 
separate binder. The archival copy is the only copy that contains the Case Report Tabulation and Case 
Report Forms. 
The Review Copy: 
The Review Copy Intended for reviewers in the corresponding technical disciplines. In addition to the 
appropriate technical section, each review copy also includes The cover letter, Form FDA-356h, 
The administrative sections, Comprehensive NDA index Individual table of contents, The Labeling 
section, and The Application Summary. 
The Field Copy:
The Field Copy required since 1993 for use by FDA inspectors during pre approval facilities inspections. It 
includes the Cover letter and Form FDA-356h, the administrative sections, the comprehensive NDA index 
Individual table of contents, The Labeling section, The Application Summary, and CMC and Methods 
Validation Package. 
NDA CONTENTS: 
The NDA may have as many as 20 different sections in addition to the Form FDA-356h itself. The specific 
contents of the NDA will depend on the Nature of the drug and the information available at the time of 
submission. The application Form FDA-356h serves as Checklist as well as Certification that the sponsor 
agrees to comply with a range of legal and regulatory requirements. 
Section 1: Index 
Section 2: Labeling 
Section 3: Application Summary 
Section 4: Chemistry, Manufacturing, and Controls (CMC) 
Section 5: Nonclinical Pharmacology and Toxicology 
Section 6: Human Pharmacokinetics and Bioavailability 
Section 7: Microbiology 
Section 8: Clinical Data 
Section 9: Safety Update Reports 
Section 10: Statistics 
Section 11: Case Report Form Tabulations 
Section 12: Case Report Forms (CRFs) 
Section 13: Patent Information 
Section 14: Patent Certification 
Section 15: Establishment Description 
Section 16: Debarment certificate 
Section 17: Field copy certification 
Section 18: User fee coversheet 
Section 19: Financial Disclosure 
Section 20: Other/ Pediatrics 
Section 1: Index: 
The NDA index is a comprehensive table of contents that enables the reviewers to find specific information 
in this massive document quickly. The NDA index should follow immediately after the Form FDA-356h 
and the administrative items. It must show the location of every section in the archival NDA by volume and 
by page number. Index It should guide reviewers the data in the technical sections, the summary, and the 
supporting documents each separately bound technical section should also contain a copy of the overall 
NDA index in addition to its own table of contents based on the index. 
Section 2: Labeling: 
The labeling section must include all draft labeling that is intended for use on the product container, 
Cartons or packages, the proposed package insert. 
Section 3: Application Summary: 
The application summary is an abbreviated version of the entire application. This overview is one of the 
few elements of the application that all reviewers receive, and it should give them a clear idea of the drug 
and its application. The summary usually comprises 50 to 200 pages. The draft product labeling include the 
following sections 
1. Description 
2. Clinical Pharmacology 
3. Indications and Usage 
4. Contraindications 
5. Warnings 
6. Precautions
7. Adverse Reactions 
8. Drug Abuse and Dependence 
9. over dosage 
10. Dosage and Administration 
11. How Supplied (primary and secondary packages) 
For each section of the labeling, include annotations referring to information in the summary and technical 
sections of the application that support the inclusion of each statement in the labeling with respect to 
Animal pharmacology and/or animal toxicology, Clinical studies, Integrated Summary of Safety (ISS) and 
Integrated Summary of Effectiveness (ISE) Safety (ISS) and Integrated Summary of Effectiveness (ISE) 
Pharmacologic class Scientific rationale Intended use Potential clinical benefits. 
Foreign marketing history: The summary must include a list of any countries in which the drug is or was 
marketed, along with the dates when it was marketed, if they are known. It must also include a list of any 
countries in which the drug has been withdrawn from marketing for any reason relating to safety or efficacy 
or in which an application has been rejected. 
Section 4: Chemistry, Manufacturing, and Controls (CMC): 
The first technical section of the NDA.It includes information on the composition, Manufacturing, and 
Specifications of the drug substance and the drug product. The CMC information must include Description 
of the drug substance or active ingredient, Its stability, Physical and chemical characteristics, Provide the 
names/designations of the drug substance, including Generic/common name Chemical name 
(IUPAC/USAN/CAS) Code(s) (CAS/internal).It Provide a structural overview including Molecular 
structure, Empirical formula, Molecular weight, Elemental composition. The description of the drug 
substance physical and chemical characteristics should include: Appearance, including color, crystalline 
form, and odor Melting/boiling point Refractive index, viscosity, and specific gravity Polymorphs, 
including modifications (forms) and relative kinetic/ thermodynamic stabilities. The physical and chemical 
characteristics should also include Solubility, Ionization constants, and Partition coefficients at various 
pHs. Solubility in common organic solvents as well as in various aqueous mediaWater0.1 N HCl 0.02 N 
HCl SGF without pepsin Water buffered to various acidic/neutral/basic pHs. It Provide a reference standard 
(RS) to elucidate the drug substance chemical structure, including Preparation method, Test methods, Test 
results as shown by a certificate of analysis (C.O.A).Provide proof that the reference standard was 
adequately tested and characterize the spectra completely. Provide structural elucidation using a reference 
standard as applicable. Measures might include X-ray (in the case of absolute configuration or 
polymorphism) UV/visible spectrum, FTIR spectrum, 1H NMR/13C NMR spectrum, Low-resolution/high-resolution 
mass spectrum, Elemental analysis. The CMC information must also include theNames, 
addresses and functions of each site where the drug substance is manufactured or tested. The description of 
the drug substance manufacturing methods must include Synthesis scheme, Synthesis description, Typical 
executed manufacturing record Compilation of and analytical controls for starting materials, Reagents, 
Solvents Catalysts, and Intermediates Suppliers for starting materials. The discussion of drug substance 
analytical controls should include the following: Specifications Methods Rationale for 
methods/specifications Method validations Batch analytical data (including impurity profiles cross-referenced 
with toxicology studies)Sampling plan Provide information on drug substance stability 
including: Ambient/accelerated stability data Retest dating Highly stressed (e.g., acid, base, reflux) data 
Provide a listing of all inactive ingredients. For compendial (e.g., UPS/NF) inactive ingredients, reference 
the appropriate current compendial monographs and provide more precise specifications as necessary. For 
noncompendial ingredients that fall under 21 CFR such as D&C and FD&C dyes, reference the appropriate 
section of 21 CFR and provide any additional specifications beyond the scope of the CFR.For 
noncompendial items that are not regulated by 21 CFR, provide appropriate analytical specifications and 
methods. Provide information on the drug product manufacturing methods: Summary and schematics of 
manufacturing procedure Master batch record for proposed marketed products, including actual operating 
conditions, type and size of equipment, and in process controls and tests Executed batch record. The section 
on drug product packaging must include: Summary of container/closure system(s)Listing of packaging
components and component/resin suppliers Specifications for each packaging component DMF 
authorization letters Description of the packaging process Test methods (as appropriate)Developmental 
data that confirms the suitability of the packaging. This includes water vapor permeation data for plastic 
containers/ closures and compatibility testing for solutions, suspensions, emulsions, etc. The drug product 
stability information will differ slightly from the drug substance stability information. Unstressed/stressed 
stability data Statistical analysis to establish consistency of data Expiration dating Post approval stability 
commitment/protocol. For an NDA, provide a list of all drug product investigational formulations used in 
clinical studies, along with the quantitative composition of each formulation. Every NDA must include an 
environmental assessment (EA)The EA, also called the environmental impact analysis report, includes an 
analysis of the manufacturing process and ultimate use of the drug product as well as a discussion of how 
the process and the drug product may affect the environment. 
Methods validation package: The final component of the CMC technical section is the methods validation 
package. The package must comprise: Specifications and test methods for each component used in the drug 
product Specifications and methods for the drug product Validation of test methods Names and addresses 
of component suppliers Names and addresses of the suppliers of the container closure system. 
Section 5: Nonclinical Pharmacology and Toxicology: 
The second technical section of the NDA provides a description of all animal and in vitro studies with the 
drug. Include a narrative summary of notable findings in all studies and a discussion of notable findings 
across the various studies. Provide individual study reports,including Pharmacology, Toxicology, and 
ADME studies. For the pharmacology studies, following data is required 
1.Effects related to the therapeutic indication, such as the pharmacodynamic ED 50 in dose-ranging studies 
and the mechanism of action (if known) 
2.Secondary pharmacological actions in order of clinical importance as possible adverse effects or as 
ancillary therapeutic effects 
3. Interactions with other drugs. 
The Toxicology information must include information on Acute toxicity, Multidose toxicity (including 
subchronic, chronic, and carcinogenicity) Special toxicity studies, as well as Reproduction studies and 
mutagenicity studies. Present toxicology data by intended route of administration in the following order: 1. 
Oral 2.Intravenous 3.Intramuscular 4.Interperitoneal 5.Subcutaneous 6.Inhalation 7 Topical 8 Other in vivo 
9.In vitro. 
Example: 
For acute toxicity studies, present the animal study data in the following order: 1. Mouse 2.Rat 3.Hamster 
4.Other rodent(s) 5.Rabbit 6.Dog 7.Monkey 8.Other nonrodent mammal(s) 9 Non mammals. 
Section 6: Human Pharmacokinetics and Bioavailability : 
This technical section includes data from Phase I safety and tolerance studies in healthy volunteers and 
ADME studies. It should include a table of PK parameters, giving the values for the major parameters 
(mean and % cv) such as Peak concentration (Cmax) Area under the curve (AUC) Time to reach peak 
concentration (tmax) Elimination constant (Ke) Distribution volume (Vd) Plasma and renal clearance 
Urinary excretion. Drug formulation information should include a list of all formulations used in clinical 
trials and in in vivo bioavailability and PK studies. The analytical methods used must be summarized in 
each in vivo biopharmaceutic study. Include detailed information, such as Sensitivity Linearity Specificity 
and Reproducibility of the analytical test methods used in each study. Provide dissolution data on each 
strength and dosage form for which an approval is sought. Include a comparative dissolution study with the 
lot in the in vivo biopharmaceutic study. Include summary of the product Dissolution performance 
Dissolution method and Dissolution specifications. 
This technical section must include individual study reports from five types of biopharmaceutic. Pilot or 
background studies Bioavailability/bioequivalence Pharmacokinetic studies Other in vivo studies In vitro 
studies . 
Section 7: Microbiology :
Required for anti infective drug products.Antimicrobial drugs differ from other classes of drugs in that they 
are designed to affect microbial physiology rather than patient physiology. In vitro and in vivo studies are 
critical in establishing the new drug effectiveness, especially if the microorganism has the potential to 
develop, or has developed, resistance to other antimicrobial drugs. This section requires the following 
technical information and data: 1. A complete description of the biochemical basis of the drug action on 
microbial physiology. 2. The drug antimicrobial spectrum. Include results of in vitro studies demonstrating 
the concentrations of the drug that are required for effective use. 3. Describe any known mechanisms of 
resistance to the drug and provide information or data of any known epidemiologic studies demonstrating 
prevalence to resistance factors. 4. Clinical microbiology laboratory methods, such as in vitro sensitivity 
discs, necessary to evaluate effective use of the drug. 
Section 8: Clinical Data: 
This technical section of the NDA comprises ten elements. The document largest and most complex 
section. List of investigators and list of INDs and NDAs Background/overview of clinical investigations 
Clinical pharmacology section Controlled clinical trials Uncontrolled clinical trials Other studies and 
information section Integrated summary of effectiveness data Integrated summary of safety information 
Drug abuse and over dosage information Integrated summary of benefits and risks of the drug 
List of investigators and list of INDs and NDAs-- The list of investigators should include all investigators 
who have used any dosage form. Alphabetize the list and note each investigator address, the type of study, 
the study identifier, and its location in the NDA .Provide a list of all known INDs under which the drug, in 
any dosage form, has been studied. 
Background/overview of clinical investigations-- Describe the general approach and rationale used in 
developing the clinical data. Explain how information about the drug derived from clinical pharmacology 
studies led to critical features of the clinical studies.Support the basis for the design features of the clinical 
trials, such as number of patients, duration, selection criteria, and controls 
Clinical pharmacology-- Clinical pharmacology Should include ADME studies, pharmacodynamic dose 
range, and dose response studies, and any other studies of the drug action. The format and order of 
presentation is as follows: 1. Table of all studies grouped by study type. Provide the investigators, study 
numbers, start date, and location of the report in the NDA. 2. For each group of studies, a brief synopsis of 
each study 3. An overall summary of the clinical pharmacology data 
Controlled clinical trials --Controlled clinical trials Provide the following material in the order presented 
below: 1. A table of all studies 2. Full clinical trial reports of all controlled studies in the following 
order: Completed studies Ongoing studies with interim results Incomplete or discontinued studies 3. 
Full reports of dose-comparison concurrent control studies, followed by those for aoeno- treatment 
concurrent control, active control studies, and historical control studies 
Uncontrolled clinical trials --Uncontrolled clinical trials They may be used to provide support for 
controlled studies and to provide critical safety information. This section should include a table of 
all studies. Group full reports of studies according to completeness and availability of Case Report 
Forms (CRFs). 
Other studies and information: Include a description and analysis of any additional information that the 
applicant has obtained from any source, foreign or domestic, that is relevant to evaluating the 
products safety and effectiveness. It should include information on commercial marketing 
experience and foreign regulatory actions, including List of countries in which the drug has been 
approved Details of any rejected registrations Copies of approved labeling (package inserts) from 
major regions such as Europe, Canada, Australia, New Zealand, and Japan Any other reports from 
the literature not provided 
Section 9: Safety Update Reports : 
A pending application must be updated when new safety data becomes available that could affect any of the 
following: Statements in draft labeling Contraindications Warnings Precautions Adverse events. 
Safety update reports are not to be used to submit any new final reports that may impact FDA 
review time unless the FDA agrees at the pre-NDA meeting that it will accept the reports in this
manner. Safety updates are submitted 4 months (120 days) after the initial application, following the 
receipt of an approval letter and at any other time that the FDA requests such an update. 
Section 10: Statistics : 
This technical section includes descriptions and documentation of the statistical analyses performed to 
evaluation the controlled clinical trials and other safety information. It must include copies of All 
controlled clinical trial reports Integrated efficacy and safety summaries Integrated summary of 
risks and benefits. 
Section 11: Case Report Form Tabulations : 
This section must include complete tabulations for each patient from every adequately or well-controlled 
Phase II and Phase III efficacy study, and from every Phase I clinical pharmacology study. It also 
must include tabulations of safety data from all clinical studies. 
Section 12: Case Report Forms (CRFs) : 
It is necessary to include the complete CRF for each patient who died during a clinical study and for any 
patients who were dropped from the study . 
The report must be submitted regardless of whether the AE is considered to be related to the study drug, 
even if the patient was receiving a placebo or comparative drug. Additional CRFs must be provided 
at the request of the FDA. 
several other supporting items as appropriate 
Item 13: Patent information 
Item 14: Patent certification 
Item15: Establishment description 
Item 16: Debarment certification 
Item 17: Field copy certification 
Item 18: User fee cover sheet (Form FDA-3397) 
Item19: Financial disclosure (Form FDA 3454, form FDA-3455) 
Item 20:Other/pediatric use. 
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 the NDA is found to be 
insufficiently complete (and reasons for this can vary from a simple administrative mistake in the 
application to a requirement to reconduct much of the testing), then the FDA rejects the application with 
the issue of a Refuse to File letter which is sent to the applicant explaining where the application has failed 
to meet requirements. Assuming that 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. A standard review implies an FDA 
decision within about 10 months while a priority review should complete within 6 months. 
The NDA in CTD Format : 
The NDA in CTD Format ICH has developed a Common Technical Document to streamline regulatory 
submissions in Europe, the U.S. and Japan. CTD is an information format that contains clinical, 
nonclinical, and manufacturing technical data.The CTD format features well-defined modules, with a 
highly specific structure and numbering of sections within the modules. It makes a clear distinction 
between subjective information sections and objective information sections. Use of the CTD format 
benefits both regulatory agencies and the pharmaceutical industry. In addition to enhancing reviews, the 
CTD use of common elements facilitates communications between the agencies and the applicants and 
simplifies the exchange of information between regulatory authorities. The document also provides a 
common basis for continuous improvement of Good Regulatory Practices. 
REFERENCES: 
Douglas J. Pisano, David S. Manlus “FDA Regulatory Affairs, A guide for Prescription Drugs, Medical 
Devices and Biologics-New drug Application “Second edition-Marcel Dekker, page no 69-108. 
http://www.fda.gov/cder/guidance/index.htm. 31
Abbreviated New Drug Application (ANDA): 
Definition : 
“A drug product that is comparable to a brand/reference listed drug product in dosage form, strength, 
route of administration, quality and performance characteristics, and intended use” 
It is termed as "abbreviated" because they generally not required to include preclinical (animal) and 
clinical (human) data to establish safety and effectiveness. 
It is an application for generic versions of off-patent drugs to receive FDA approval with less costly or no 
preclinical and clinical testing. 
The intention is to minimize duplication of available information. No safety or efficacy trials are 
performed. Complete chemistry, manufacturing, and controls information and information regarding the 
bioavailability of solid dosage forms. 
Basic Generic Drug Requirements are:-- 
· Same active ingredient(s) 
· Same route of administration 
· Same dosage form 
· Same strength 
· Same conditions of use 
· Inactive ingredients already approved in a similar NDA 
Goal of ANDA: 
Provide enough information to permit FDA to make the following key decisions: 
· Whether the drug is safe and effective in its proposed use(s), and whether the benefits of the 
drug outweigh the risks. 
· Whether the drug's proposed labeling (package insert) is appropriate, and what it should 
contain. 
· Whether the methods used in manufacturing the drug and the controls used to maintain the 
drug's quality are adequate to preserve the drug's identity, strength, quality, and purity. 
· To reduce the price of the drug. 
· To reduce the time development. 
· Increase the bioavailability of the drug in comparison to references list drug. 
Abbreviated or Supplemental NDA: 
· Abbreviated NDA: includes.. 
· Generic drug 
· New combination of approved drugs 
· Proportion of ingredients in combination is changed 
· Supplemental NDA: includes.. 
· New intended use of the drug (labeling change) 
· Dose, method or duration of administration is changed 
· Change in manufacturing process or location 
NDA vs. ANDA Review Process: 
Requirements for NDA: 
· Labeling 
· Pharmacology and toxicology 
· Chemistry 
· Manufacturing 
· Controls 
· Microbiology
· Inspection 
· Testing 
· Animal studies 
· Human studies 
· Bioavailability 
Requirements for ANDA: 
· Labeling 
· Pharmacology and toxicology 
· Chemistry 
· Manufacturing 
· Controls 
· Microbiology 
· Inspection 
· Testing 
· Bioequivalence 
NDA ANDA 
Applicable for new drug Applicable for generic drug 
Takes longer time ( 12-15 years) Comparatively less time taken(1-2 years) 
More expenditure of money Comparatively less 
Cost of drugs are more Cost of drugs are less 
Nonclinical studies and clinical 
investigations are essential 
Nonclinical studies and clinical investigations 
are nonessential except bioavailability and 
bioequivalence 
Format of ANDA: 
Three copies of application are required, an archival copy, a review copy and a field copy. FDA will 
maintain guidance documents on the format and content of applications to assist applicants in their 
preparation. ANDA includes the following: 
· Application form: the applicant shall submit a completed and signed 
application form that contains the information. The applicant shall state whether the submission is an 
abbreviated application or a supplement to an abbreviated application. 
· Table of contents: the archival copy of the ANDA is required to contain a 
table of contents that shows the volume number and page number of the contents of the submission. 
· Basis for ANDA submission: an ANDA must refer to a listed drug. 
Ordinarily, that listed drug will be the drug product selected by the agency as the reference standard for 
conducting bioequivalence testing. The application shall contain: 
· The name of the reference drug, including its dosage from and strength. 
For an abbreviated new drug application based on the reference listed drug must be the same as the 
listed drug approved in the petition. 
· A statement as to whether, according to the information published in the 
list, the reference listed drug is entitled to a period of marketing exclusivity. 
· Conditions of use: 
· A statement that the conditions of use prescribed, recommended, or suggested in the labeling 
proposed for the drug product have been previously approved for the reference listed drug. 
· A reference to the applicant’s annotated proposed labeling and to the currently approved labeling 
for the reference listed drug.
· Active ingredients 
· Route of administration, dosage form and strength. 
· Bioequivalence 
· Labeling 
· Samples: need not be submitted until requested by FDA. 
· Patent certification 
· Financial certification or disclosure statement. 
ANDA contents: 
Section 1: Overall ANDA index:- The NDA index is a comprehensive table of contents that enables the 
reviewers to find specific information in this massive document quickly. 
Section 2: Labeling:- It must include all draft labeling that is intended for use on the product container, 
cartons or packages, including the proposed package insert. 
Section 3: Application summary:- Proposed annotated package insert- 
· Pharmacology class, scientific rational, intended use, and potential clinical 
benefits 
· Foreign marketing history 
· Chemistry, Manufacturing and control summary 
· Nonclinical pharmacology and toxicology summary 
· Human pharmacokinetics and bioavailability summary 
· Microbiology summary 
· Clinical data summary and results of statistical analysis 
· Discussion of benefit/risk relationship 
Section 4: Chemistry, manufacturing and controls:- 
· Chemistry, manufacturing and control information 
· Samples 
· Methods validation package 
Section 5: Nonclinical pharmacology and toxicology 
· Provide individual study reports, including pharmacology, toxicology, 
ADME studies. 
· Effects related to the therapeutic indication, such as the pharmacodynamic 
ED50 in dose- ranging studies and the mechanism of act ion (if know n) 
· Interactions with other drugs (or cross-reference the location of the 
information in any of the above subsection 
Section 6: Human Pharmacokinetics and bioavailability:- includes data from Phase I safety and 
tolerance studies in healthy volunteers. Element in the section tabulated summary of studies showing all in 
vivo biopharmaceutics studies performed. 
· Summary of analytical method used in in vivo biopharmaceutic study 
· Pilot or background studies 
· Bioavailibility or bioequivalence studies 
· Pharmacokinetic studies 
· In vitro studies 
Section 7: Microbiology:- Includes for anti infective drug products. It requires the following technical 
information and data:- 
· A complete description of the biochemical basis of the drug action on 
microbial physiology 
· The drugs antimicrobial spectrum
· Describe any known mechanism of resistance to the drug and provide 
information/data of any known epidemiologic studies demonstrating prevalence to resistance factor 
· Clinical microbiology laboratory methods 
Section 8: Safety data 
·· 
Statements in draft labeling 
· Contra indications 
· Warnings 
· Precautions 
· Adverse events 
Section 9: Statistical data 
· All controlled clinical trial reports 
· Integrated efficacy and safety summaries 
· Integrated summary of risks and benefits 
Section 10: Case report tabulation:- Includes complete tabulation for each patient from every adequately 
are well controlled phase II and Phase III efficacy, clinical pharmacology study. It also tabulation of safety 
data from all clinical studies. 
Section 11: Case report forms:- Includes the complete CRF for each patient who died during a clinical 
study or adverse event, regardless of whether the AE is considered to be related to the study drug, even if 
the patient was receiving a placebo or comparative drug. 
Bioequivalence: 
A generic drug is considered to be bioequivalent to the brand name drug if: 
· The rate and extent of absorption do not show a significant difference from listed drug, or 
· The extent of absorption does not show a significant difference and any difference in rate is 
intentional or not medically significant. 
Bio equivalent Inequivalent 
Resources for ANDA Submissions:- The following resources have been gathered to provide you with the 
legal requirements of an ANDA application, assistance from CDER to help you meet those requirements, 
and internal ANDA review principles, policies and procedures. 
Guidance Documents for ANDAs:- Guidance documents represent the Agency's current thinking on a 
particular subject. These documents are prepared for FDA review staff and applicants/sponsors to provide 
guidelines to the processing, content, and evaluation/approval of applications and also to the design, 
production, manufacturing, and testing of regulated products. They also establish policies intended to 
achieve consistency in the Agency's regulatory approach and establish inspection and enforcement 
procedures. Because guidances are not regulations or laws, they are not enforceable, either through 
administrative actions or through the courts. An alternative approach may be used if such an approach 
satisfies the requirements of the applicable statute, regulations, or both. For information on a specific 
guidance document, please contact the originating office. They includes… 
· Generics
· Generics (Draft - Distributed for comment purposes only) . 
· Procedural Draft: Applications Covered by Section 505(b)(2) (Issued 10/1999, 
Posted 12/7/1999). This provision permits FDA to rely, for approval of an NDA, on data 
not developed by the applicant. 
· Biopharmaceutics. 
· Bioavailability and Bioequivalence Studies for Orally Administered Drug Products - 
General Considerations (Issued 10/2000, Posted 10/27/2000). This guidance should be 
useful for applicants planning to conduct bioavailability (BA) and bioequivalence (BE) 
studies during the IND period for an NDA, BE studies intended for submission in an 
ANDA, and BE studies conducted in the post-approval period for certain changes in both 
NDAs and ANDAs. 
· Drug Master Files. A Drug Master File (DMF) is a submission to the FDA that may be 
used to provide confidential detailed information about facilities, processes, or articles used in the 
manufacturing, processing, packaging, and storing of one or more human drugs. 
· Required Specifications for FDA's IND, NDA, and ANDA Drug Master File Binders 
· Guidance for Industry: Changes to an Approved NDA or ANDA 
· Refusal to Receive. (Issued 7/12/1993, Posted 11/26/99) Clarifies CDER's decisions to 
refuse to receive an incomplete application. 
· Inactive Ingredient Database. This database contains all inactive ingredients present in 
approved drug products or conditionally approved drug products currently marketed for human use. 
Laws, Regulations, Policies and Procedures: 
The mission of FDA is to enforce laws enacted by the U.S. Congress and regulations established by the 
Agency to protect the consumer's health, safety, and pocketbook. The Federal Food, Drug, and Cosmetic 
Act is the basic food and drug law of the U.S. With numerous amendments it is the most extensive law of 
its kind in the world. The law is intended to assure consumers that foods are pure and wholesome, safe to 
eat, and produced under sanitary conditions; that drugs and devices are safe and effective for their intended 
uses; that cosmetics are safe and made from appropriate ingredients; and that all labeling and packaging is 
truthful, informative, and not deceptive. 
Code of Federal Regulations (CFR): 
The final regulations published in the Federal Register16 (daily published record of proposed rules, final 
rules, meeting notices, etc.) are collected in the CFR. The CFR is divided into 50 titles which represent 
broad areas subject to Federal regulations. 
The FDA's portion of the CFR interprets the Federal Food, Drug and Cosmetic Act and related statutes. 
Section 21 of the CFR contains most of the regulations pertaining to food and drugs. The regulations 
document most actions of all drug sponsors that are required under Federal law. The following regulations 
apply to the ANDA process: 
· (21CFR Part 314) Applications for FDA Approval to Market a New Drug or and 
Antibiotic Drug 
· (21CFR Part 320) Bioavailability and Bioequivalence Requirements. For more 
information on retention samples, please see Bioequivalence Study Retention Samples. 
· Bioavailability and Bioequivalence Requirements; Abbreviated Applications; 
Final Rule. (Issued and posted 12/19/2002) 
· (21CFR Part 310) New Drugs 
MaPPs :- CDER's Manual of Policies and Procedures (MaPPs) provide official instructions for internal 
practices and procedures followed by CDER staff to help standardize the drug review process and other 
activities, both internal and external. MaPPs define external activities as well. 
ANDA review process:
Time frames for reviewing ANDA by FDA: 
Within 180 days of receipt of an application for a new drug under section 505(b) of the act or an 
abbreviated application for a new drug under section 505(j) of the act, FDA will review it and send the 
applicant either an approval letter under 314.105 or a complete response letter under 314.110. This 180-day 
period is called the "initial review cycle." 
At any time before approval, an applicant may withdraw an application under 314.65 or an abbreviated 
application under 314.99 and later submit it again for consideration. 
The initial review cycle may be adjusted by mutual agreement between FDA and an applicant or as 
provided in 314.60 and 314.96, as the result of a major amendment. 
Filing of ANDA: 
Within 60 days after FDA receives an application, the agency will determine whether the application may 
be filed. The filing of an application means that FDA has made a threshold determination that the 
application is sufficiently complete to permit a substantive review. If FDA finds that none of the reasons 
for refusing to file the application apply, the agency will file the application and notify the applicant in 
writing. The date of filing will be the date 60 days after the date FDA received the application. The date of
filing begins the 180-day period described in section 505(c) of the act. This 180-day period is called the 
"filing clock." 
Approval of ANDA: 
FDA will approve an application and issue the applicant an approval letter on the basis of draft labeling if 
the only deficiencies in the application concern editorial or similar minor deficiencies in the draft labeling. 
Such approval will be conditioned upon the applicant incorporating the specified labeling changes exactly 
as directed, and upon the applicant submitting to FDA a copy of the final printed labeling prior to 
marketing. 
Amendments to an unapproved ANDA: 
· An applicant may amend an abbreviated new drug application that is submitted, but not yet 
approved, to revise existing information or provide additional information. Amendments containing 
bioequivalence studies must contain reports of all bioequivalence studies conducted by the applicant on 
the same drug product formulation, unless the information has previously been submitted to FDA in the 
abbreviated new drug application. A complete study report must be submitted for any bioequivalence 
study upon which the applicant relies for approval. For all other bioequivalence studies conducted on 
the same drug product formulation, the applicant must submit either a complete or summary report. If a 
summary report of a bioequivalence study is submitted and FDA determines that there may be 
bioequivalence issues or concerns with the product, FDA may require that the applicant submit a 
complete report of the bioequivalence study to FDA. 
· Submission of an amendment containing significant data or information before the end of the initial 
review cycle constitutes an agreement between FDA and the applicant to extend the initial review cycle 
only for the time necessary to review the significant data or information and for no more than 180 days. 
· The applicant shall submit a field copy of each amendment. The applicant, other than a foreign 
applicant, shall include in its submission of each such amendment to FDA a statement certifying that a 
field copy of the amendment has been sent to the applicant's home FDA district office. 
References: 
· Richard A. Guarino- New Drug Approval process-1)The New Drug Application, Content, Format 
2) Abbreviated $ Supplementary New Drug Application- Fourth edition-Marcel Dekker,inc- page no 
113-183. 
· Douglas J. Pisano, David S. Manlus –FDA Regulatory Affairs, A guide for Prescription Drugs, 
Medical Devices and Biologics-New drug Application –Second edition-Marcel Dekker,inc- page no 
69-108. 
· Loyd V. Allen Jr, Nicholas G. Popovich, Howard C. Ansel’s Pharmaceutical Dosage Forms and 
delivers systems- New Drug Development and Approval Process-8th edition- B.I. publication- Page no 
25-65. 
· http://www.fda.gov/cder/guidance/index.htm . 
· http://www.fda.gov/drugs/developmentapprovalprocess/howdrugsaredevelopedandapproved/approv 
alapplications/abbreviatednewdrugapplicationandagenerics/default.htm 
· http://www1.pointcross.com/source/ddg/charts/andachart.htm 
· http://www.accessdata.fda.gov/script s /cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=314.94 
Common technical document: 
The common technical document was agreed in November 2000 in sandicgo USA. It provides for a 
harmonized structure and format for new product applications. This is one in a series of guidance that 
provide recommendations for applicants preparing the CTD for the registration of pharmaceuticals used for 
human use, that is , submission to the USFDA . It will significantly reduce time resources. This applicant 
have used many different approaches to organize the information, but differences in organization of each 
application made regulatory agency review, more difficult and may lead to omission of critical data or 
analysis. Such omission can result in a decision by regulatory agency which is not correct for a given drug.
So, as to overcome this “US federal drug administration ensured that the worldwide pharmaceutical 
industry should follow tightly regulated strict standard document maintained in a common format from 
initial project definition to product approval. 
· In addition exchange of regulatory information among regulatory authorities 
will be simplified. 
· The FDA characterized the CTD as “ an information package of clinical, non clinical and 
manufacturing, technical data in the same format and with the same content , that would be 
submitted for registering new drugs in all three ICH the US, European union of Japan. 
· European Union has its own requirements for the organization of technical reports in 
submission and for the preparation of the summaries and tables. 
· In Japan the applicants must prepare the GAIYO which organizes and presents a summary of 
the technical information. 
· In Europe experts reports and tabulated summaries are required and return summaries are 
recommended. 
· In US FDA has guidance regarding the format and content of new drug application 
submission. 
Guidance for industry on preparing the CTD: 
It has been divided into 4 guidance document on harmonized format. 
· The organization of the CTD 
· The quality section 
· The efficacy section 
· The safety section 
Human use and has been subject to consultation by the regulatory parties in accordance with the ICH process. 
· General considerations for submitting marketing applications according to ICH/CTD format. 
· CTD guidance’s are intended to be used together with other ICH and agency guidance’s 
· The guidance suggest that controlled procedure need to be implemented, to specific address – 
maintenance, storage, access restriction, technical approach, and responsibilities of personal relevant 
tasks across the retention period of an electronic record. 
Preparing of organizing the CTD: 
· The guidance address the general organization of the information to be presented in CTD 
application for new pharmaceuticals(including biotechnology derived products) 
· Guidance’s also are available that discuss the quality, efficacy and safety section of the CTD 
in respective modules. 
· Applicants should not modify the overall origination of the CTD 
However in the non clinical and clinical summarizes section of the CTD, applicants can 
modify individual formats, to provide the best possible presentation of the technical 
information and to facilitate the understanding evaluation of the results 
Throughout the CTD, the display of information should be unambiguous and transparent to 
facilitate the review of the basic data and to help a reviewer become quickly oriented to 
application contents 
· Text and tables should be prepared using margins’ that allow the document to be 
printed on both A4 paper(EU and Japan ) 8.5×1111 paper(US) 
· The left hand margin should be sufficiently large that information is not obscured 
through binding. 
· Font sizes for text and tables should be of a style and size that are large enough to be 
easily legible, even after photocopying. 
· 12. Point Font is required for narrative text
· Acronyms and abbreviations should be defined the first time they are used in each 
module. 
The Common Technical Document (CTD) is a set of specification for application dossier for she 
registration of Medicines and designed to be used across Europe, Japan and the United States. It was 
developed by the European Medicines Agency (EMEA, Europe), the Food and Drug Administration (FDA, 
U.S.) and the Ministry of Health, Labor and Welfare (Japan). The CTD is maintained by the International 
Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human 
Use (ICH).[1] 
The Common Technical Document is divided into five modules: 
Administrative and prescribing information 
Overview and summary of modules 3 to 5 
Quality (pharmaceutical documentation) 
Safety (toxicology studies) 
Efficacy (clinical studies) 
Detailed subheadings for each Module are specified for all jurisdictions. The contents of Module 1 and 
certain subheadings of other Modules will differ, based on national requirements. 
After the United States, European Union and Japan, the CTD has been adopted by several other countries 
including Canada and Switzerland. 
The Paper CTD is destined to be replaced by its electronic counterpart, the eCTD. 
Organisation of the Common Technical Document 
The Common Technical Document is organized into five modules. Module 1 is region specific. Modules 
2, 3, 4, and 5 are intended to be common for all regions. Conformance with this guideline should ensure 
that these four modules are provided in a format acceptable to the regulatory authorities. 
Module 1. Administrative Information and Prescribing Information 
This module should contain documents specific to each region; for example, application forms or the 
proposed label for use in the region. The content and format of this module can be specified by the relevant 
regulatory authorities. 
Module 2. Common Technical Document Summaries 
Module 2 should begin with a general introduction to the pharmaceutical, including its 
pharmacologic class, mode of action, and proposed clinical use. In general, the Introduction should not 
exceed one page. 
Module 2 should contain 7 sections in the following order: 
CTD Table of Contents 
CTD Introduction
Quality Overall Summary 
Nonclinical Overview 
Clinical Overview 
Nonclinical Written and Tabulated Summaries 
Clinical Summary 
The organisation of these summaries is described in Guidelines for M4Q, M4S, and M4E. 
M4-Q: the CTD for the registration of pharmaceuticals for human use –quality 
·The quality section of the CTD provides a harmonized structure and format for presenting CMC 
(chemistry, manufacturing, controls) information in a registration dossier. 
·The table of contents include sections on drug substance and drug product. 
·Due to the fact that many CMC topics have not yet been the subject of ICH guidelines (e.g.: drug 
substance synthesis, drug product manufacture), contain the content of CTD-Q is not totally 
harmonised. 
·A new section on pharmaceutical development and also a new CMC summary document, the 
quality overall summary has been developed. 
M4-S: the CTD for registration of pharmaceuticals for human use- safety 
Non –clinical summaries and organisation of module-4 
· The M4S guideline will outline the structure of format of nonclinical summaries in module2 
of the CTD and will provide the organisation of module 4 i.e. the non-clinical study reports. 
· The non-clinical overview should present an should present an integrated and critical 
assessment of the pharmacological, pharmacokinetic, and toxicological evolution of the 
pharmaceutical and generally not to exceed 30 pages. 
· The non-clinical written summaries (100-150 pages) are to provide more extensive summaries 
and discussion of non clinical information on pharmacological, pharmacokinetics and toxicology. 
M4-E: the CTD for the registration of pharmaceuticals for human use-efficacy 
Module-2:clinical overview and clinical summary 
Module -5:clinical study reports. 
M4E describes the structure and format of the clinical data in an application, including summaries and 
detailed study reports. 
There are two high level clinical summaries in module 2 of the CTD: 
Clinical overview: a short document that provides a critical assessment of the clinical data 
Clinical summary: a longer document that focuses on data summarisation and integration. 
Module 3. Quality 
Information on Quality should be presented in the structured format described in Guideline M4Q. 
Module 4. Nonclinical Study Reports 
The nonclinical study reports should be presented in the order described in Guideline M4S. 
Module 5. Clinical Study Reports 
The human study reports and related information should be presented in the order described in Guideline 
M4E. 
Organisation of the Common Technical Document for the 
Registration of Pharmaceuticals for Human Use 
Module 1: Administrative Information and Prescribing Information 
Table of Contents of the Submission Including Module 1 
Documents Specific to Each Region (for example, application forms, prescribing information) 
Module 2: Common Technical Document Summaries 
Common Technical Document Table of Contents (Modules 2-5) 
CTD Introduction
Quality Overall Summary 
Nonclinical Overview 
Clinical Overview 
Nonclinical Written and Tabulated Summaries 
Pharmacology 
Pharmacokinetics 
Toxicology 
Clinical Summary 
Biopharmaceutical Studies and Associated Analytical Methods 
Clinical Pharmacology Studies 
Clinical Efficacy 
Clinical Safety 
Literature References 
Synopses of Individual Studies 
Module 3: Quality 
Table of Contents of Module 3 
Body of Data 
Literature References 
Module 4: Nonclinical Study Reports 
Table of Contents of Module 4 
Study Reports 
Literature References 
Module 5: Clinical Study Reports 
Table of Contents of Module 5 
Tabular Listing of All Clinical Studies 
Clinical Study Reports 
Literature References 
Reference: 
·Guidance for Industry, ICH M4: Organization of the CTD" U.S. Department of Health and Human 
Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center 
for Biologics Evaluation and Research (CBER) August 2001 
SUPAC 
( SCALE - UP AND POST- APPROVAL CHANGES ) 
Product development aims at formulating active drug ingredient in a palatable form . 
Technology transfer of a pharmaceutical product from research to the production floor (shop floor) with 
simultaneous increase in production outputs is commonly known as scaleup.In simple terms the process of 
increasing batch size is termed as scaleup.Scale down refers to decrease in batch size in response to 
reduced market requirements. SUPAC refers to the FDA recommended testing and filling actions to be 
taken by a pharmaceutical firm when it changes the manufacturing process of a drug product that has been 
approved via a New Drug Application(NDA),an Abbreviated New Drug Application (ANDA),or an 
Abbreviated Antibiotic Drug Application(AADA). It is designed to lower the regulatory burden associated 
with alterations in batch size ,components and composition ,manufacturing site or manufacturing of a 
marketed product. 
A drug may experience many changes during its life cycle .These changes may have an adverse effect on 
the overall safety and effectiveness of the drug or drug product .After a number of changes over a long time 
period ,the product coming to market maybe completely different from the one that was approved.Hence 
data submitted to regulatory authorities in support of a change must have a comparison record of the drug 
or drug product to the one that was approved initially.documentation generated in support of any change to
the approved drug is submitted to regulatory authority for review,and based on the benefit to risk ratio ,the 
drug or drug product is approved.depending upon the intensity of change ,supporting documents are 
provided to the regulatory agency. 
The SUPAC Guidance offer an advantage over the regulations that the documents are specific for particular 
dosage forms.This approach was taken since some product types are more complicated than others,and 
require more complicated controls.Two guidance have been finalized.They are : 
1. Immediate release solid oral dosage forms –Scale up and post approval changes 
:Chemistry,Manufacturing and controls,in vitro dissolution testing ,and invivo bioequivalence 
documentation. 
2. Non sterile semisolid dosage forms –Scaleup and post approval changes : :Chemistry, 
Manufacturing and controls, in vitro dissolution testing ,and invivo bioequivalence documentation. 
In addition ,SUPAC documents covering other dosage forms (eg, extended-release 
products,transdermals,parenteral solutions)as well as a related documents for bulk active substances ,are at 
various stages of development. 
Modifications are divided into one of three categories, each with specific testing and documentation 
requirements. 
Level 1 changes: Are those that are unlikely to have any detectable impact on formulation quality or 
performance ,such as change of less than 5% in the concentration of an excipient. 
likely to have a significant impact on quality or performance. 
SUPAC Stability Testing: 
In addition to in vitro release testing ,longterm stability studies are required at all levels of change ,while 
accelerated stability studies are required for levels 2 and 3. 
SUPAC Testing : 
The purpose of SUPAC Testing is to verify that there is no change in product quality or performance after 
a modification .A key component of a level 2 or 3 change is in vitro release testing for pre and post change 
formulations. 
For SUPAC Submissions,the following points should also be considered : 
1.In most cases, except those involving scale-up,stability data from pilot scale batches will be acceptable to 
support the proposed change. 
2.Where stability data show a trend towards potency loss or degradant increase under accelerated 
conditions ,it is recommended that historical accelerated stability data from a representative prechange 
batch be submitted for comparision. 
An invitro release rate can reflect the combined effect of several physical and chemical parameters 
,including solubility and particle size of the active ingredient and rheological properties of the 
dosageform.In most cases,invitro release rate is a useful test assess between prechange and post change 
products. 
1.In vitro release testing is a useful test to assess product “sameness”under certain scaleup and post 
approval changes for semisolid products. 
2.The development and validation of an invitro release test are not required for approval of an 
NDA,ANDA,AADA nor is the invitro release test required as a routine batch to batch quality control test. 
3.In vitro release testing ,alone ,is not a surrogate test for in vivo bioavailability or bioequivalence. 
4.The in vitro release rate shouldnot be used for comparing different formulations across manufacturers. 
The SUPAC guidance documents defines 
1)levels of change 
2)recommended chemistry,manufacturing,and controls tests for each level of change. 
3)In vitro dissolution tests and in vivo bioequivalence tests for each level of change. 
4)documentation that should support the change for new drug applications(NDA)and abbreviated new drug 
applications(ANDAs).
This guidance provides recommendations to sponsors of new drug applications(NDA),abbreviated 
antibiotic applications(AADA’s),who intend during the post approval period,to change :1)The components 
or composition:2)the site of manufacture3)The scale up/scale down of manufacture 4)the 
manufacturing(process and equipment)of an immediate release oral formulations. 
The purpose of this guidance is to provided recommendations to pharmaceutical Manufacturers using the 
Center for Drug Evaluation and Research’s guidance for Industry: Immediate Release Solid Oral Dosage 
Forms-Scale up and post approval changes: Chemistry, manufacturing and controls ,In vitro Dissolution 
testing ,and in vivo Bioequivalence Documentation(SUPAC-IR) ,which is published in November 1995, 
and guidance for industry. 
Dissolution testing: Case A: Dissolution of Q=85%in 15min in 900milliliters of 0.1N hydrochloride using 
the United States Pharmacopoeia. Apparatus 1 at 100 revolutions per minute or Apparatus 2 At 50 rpm. 
Case B: Multi point dissolution profile in the application/compendial medium at 15,30,45,60,120min until 
an asymptote is reached for the proposed and currently Accepted formulation. 
Case C: Multi point dissolution profiles performed in water ,0.1N HCL,and USP buffer media at pH 
4.5,6.5,7.5 for the proposed and currently accepted formulations. Adequate sampling should be performed 
at 15,30,45,60,120 min until either 90% of drug from the drug product is dissolved or an asymptote is 
reached .A surfactant maybe used with appropriate justification. 
Components and composition: 
It focuses on changes in excipients in the drug product Changes in the amount of drug substance are not 
addressed by this guidance Changes in components or composition that have the effect of adding a new 
excipient or deleting an excipient. 
A.Level 1 changes. 
1.Definition of level 
Level 1 changes are those that are unlikely to have any detectable impacts on formulation quality and 
performance. 
Examples: 
a.Deletion or partial deletion of an ingredient intended to affect the color or flavor of the drug product ;or 
change in the ingredient of the printing ink to another approved ingredient . 
b.changes in excipients ,expressed as percentage (w/w)of total formulation ,less than or equal to the 
following percent ranges: 
Excipient percent excipient(w/w)out of total target dosage form weight 
Filler ± 10 
Disintigrant-starch ±6 : other ± 2 
Binder ± 1 
Lubricant –calcium or magnesium stearate ± 0.5 
Other ± 2 
Glidant - talc ±2 
other±0.2 
filmcoat- ±2 
These percentages are based on the assumption that the drug substance in the drug product is formulated to 
100%of label /potency .The total additive effect of all excipient changes should not change by more than 
10%. 
2. Test documentation 
a. Chemistry documentation: 
Application /compendial release requirements and batch records. 
Stability testing:one batch on long term stability data reported in annual report. 
b.Dissolution documentation 
c.Invivo Bioequivalence documentation
3.Filing documentation-prior approval supplement(all information including accelerated stability 
data);annual report(long term stability data). 
Level 2 changes: are those that could have a significant impact on formulation quality and performance. 
Tests and filing documentation for a level 2 change vary depending on three factors :therapeutic 
range,solubility,permeability. 
Level 3 changes:are those that are likely to have a significant impact on formulation quality and 
performance.Tests and filling documentation vary depending on the 3 factors;therapeutic range 
,solubility,permeability.. 
SITE CHANGES: 
Site changes consist of changes in location of the site of manufacture for both company-owned and contract 
manufacturing facilities and donot include any scaleup changes ,changes in manufacturing(including 
process and/or equipment),or changes in components or composition .New manufacturing locations should 
have a satisfactory current Good manufacturing practice (CGMP)inspection.. 
A.Level 1 changes 
1.Definition of level 
Level 1 changes consists of site changes with in a single facility where the same equipment ,standard 
operating procedures ,environmental conditions sites are used,and where no changes are made.and controls 
and personnel common to both manufacturing to the manufacturing batch records except for administrative 
information and the location of the facility.common is defined as employees already working on the 
campus who have suitable experience with the manufacturing process. 
2.Test documentation 
a)Chemistry documentation 
None beyond application/compendial release requirements. 
b)Dissolution documentation 
None beyond application/compendial release requirements. 
c)Invivo bioequivalence documentation 
None 
3.Filing documentation 
Annual report 
B.Level 2 changes 
1.Definition of level 
Level 2 changes consists of site changes with in a contiguous campus,or between facilities in adjacent city 
blocks,where the same equipment,SOP’s,environmental conditions(eg temparature and humidity)and 
controls,and personnel common to both manufacturing sites are used ,and where no changes are made to 
the manufacturing sites are used ,and where no changes are made to the manufacturing batch records,except 
for administrative information and the location of the facility. 
2.Test documentation 
a.Chemistry Documentation 
Location of new site and updated batch records.None beyond application /compendial release requirements. 
One batch on long term stability data reported in annual report. 
b.Dissolution Documentation 
None beyond application /compendial release requirements. 
c.Invivo Bioequivalence documentation 
None 
3.Filing documentation 
Changes being effected supplement :annual report(long term stability test data) 
C.Level 3 changes 
1.Definition of level
Level 3 changes consists of a change in manufacturing site to a different campus.A different campus is 
defined as one that is not on the same original contiguous site or where the facilities are not in adjacent city 
blocks.To qualifyas a level 3 change,The same equipment ,SOP’s ,environmental conditions ,and controls 
shouldbe used in the manufacturing process at the new site ,and no changes maybe made to the 
manufacturing batch records except for administrative information ,location,language translation,where 
needed. 
2.Test documentation 
a)Chemistry documentation 
Location of new site and updated batch records. 
Application/compendial release requirements . 
Stability: 
Significant body of the data available: 
One batch with three months acclerated stability data reported in supplement;one batch on longterm 
stability data reported in annual report. 
Significant body of the data not available: 
Up to three batches with three months acclerated stability data reported in supplement;up to three batches 
on long term stability data reported in annual report. 
b)Dissolution documentation 
CaseB:multi point dissolution profile should be performed in the application/compendial medium at 
15,30,45,60,120 minutes or until an asymptote is reached. 
The dissolution profile of the drug product at the current and proposed site should be similar. 
c) Invivo Bioequivalence Documentation: 
None 
3. Filing Documentation 
Changes being effected supplement;annual report(long term stability data). 
CHANGES IN BATCH SIZE (SCALE- UP/SCALE DOWN) : 
Post approval changes in the size of a batch from the pivotal/pilot scale batch material to larger or smaller 
production batches call for submission of additional information in the application. Scale down below 
100,000 dosage units is not covered by this guidance .All scale-up changes should be properly validated 
and, where needed, inspected by appropriate agency personnel. 
A)Level 1 changes 
1. Definition of level 
Change in batch size up to and including a factor of 10 times the size of the pilotbio batch where; 
i) The equipment used to produce the test batches is of the same design and operating principles; 
ii) The batches is manufactured in full compliance with CGMP’s. 
iii) The same standard operating procedure(SOP’s)and controls, as well as the same formulation and 
manufacturing procedures, are used on the test batches and on the full scale production batches 
2. Test documentation 
a)Chemistry documentation 
Application/Compendial release requirements .Notification of change and submission of updated batch 
records in annual report. 
One batch on long term stability reported in annual report . 
b)Dissolution documentation 
None beyond application /compendial release requirements. 
c)In vivo Bio equivalence 
None 
3.Filing Documentation 
Annual report (long term stability data). 
B)Level 2 changes
1)Definition of level 
Changes in batch size beyond a factor of ten times the size of the pilot/biobatch,where; 
i)The equipment used to produce the test batches is of the same design and operating principles; 
ii)The batches is manufactured in full compliance with CGMP’s. 
iii)The same standard operating procedure(SOP’s)and controls, as well as the same formulation and 
manufacturing procedures,are used on the test batches and on the full scale production batches. 
2.Test documentation 
a)Chemistry documentation 
Application/compendial release requirements .Notification of change and submission of updated batch 
records. 
Stability testing: 
One batch with three months accelerated stability data and one batch on longterm stability. 
b)Dissolution documentation 
Case B testing 
c)In vivo Bioequivalence documentation 
None. 
3.Filing Documentation 
Changes being effected supplement :annual report(long term stability data) 
MANUFACTURING CHANGES: 
Manufacturing changes may affect both equipment used in the manufacturing process and the process 
itself. 
A.Equipment 
1.Level 1 changes 
a)Definition of change 
This category consists of : 
1)Change from non-automated or non mechanical equipment to move ingredients:and 
2)Change to alternative equipment of the same design and operating principles of the same or of a different 
capacity. 
b)Test documentation 
i)Chemistry documentation 
Application/compendial release requirements .Notification of change and submission of updated batch 
records. 
Stability testng:One batch on longterm stability. 
b)Dissolution documentation 
None beyond application/compendial release requirements. 
c)In vivo Bioequivalence documentation 
None. 
c)Filing documentation 
Annual report(Long term stability data 
2.Level 2 Changes 
a)Definition of level 
Change in equipment to a different design and different operating principles. 
b)Test documentation 
i)Chemistry documentation 
Application/compendial release requirements .Notification of change and submission of updated batch 
records. 
Stability testing 
Significant body of data available: 
One batch with three months accelerated stability data reported in supplement ;One batch on long term 
stability data reported in annual report.
Significant body of data not available 
Up to three batches with three months accelerated stability data reported in supplement;upto three batches 
on long term stability data reported in annual report. 
ii)Dissolution documentation 
Case C dissolution profile. 
iii)Invivo Bioequivalence documentation 
None 
c.Filing documentation 
Prior approval supplement with justification for change ;annual report(long term stability data). 
B)Process 
a)Definition of level 
This category includes process changes including changes such as mixing times and operating speeds 
within applicaton/validation ranges. 
b)Test documentation 
i)Chemistry documentation 
None beyond application/compendial release requirements. 
ii)Dissolution documentation 
None beyond application/compendial release requirements 
iii)In vivo Bioequivalence documentation 
None 
c).Filing documentation 
Annual report 
2.Level 2 changes 
a.Definition of level 
This category includes process changes including changes such as mixing times and operating speeds 
outside of application / validation ranges. 
b)Test documentation 
i)Chemistry documentation 
Application/compendial release requirements .Notification of change and submission of updated batch 
records. 
Stability testing:one batch on longterm stability. 
ii)Dissolution documentation 
Case B dissolution profile. 
iii)Invivo Bioequivalence documentation 
None 
c).Filing documentation 
Changes being effected supplement ;annual report(long term stability data). 
3.Level 3 changes 
a).Definition of level 
This category includes changes in the type of process used in the manufacture of the product ,such as a 
change from wet granulation to direct compression of dry powder. 
b)Test documentation 
i)Chemistry documentation 
Application/compendial release requirements .Notification of change and submission of updated batch 
records. 
Stability testing: 
Siignificant body of data available: 
One batch with three months accelerated stability data reported in supplement ;one batch on long term 
stability data reported in annual report. 
Significant body of data not available:
Up to three batches with three months accelerated stability data reported in supplement ;up to three batches 
on long term stability data reported in annual report. 
ii)Dissolution documentation 
case B dissolution. 
iii)Invivo Bioequivalence documentation 
Invivo bioequivalence study .The bioequivalence study maybe waived if a suitable in vivo/invitro 
correlation has been verified. 
c).Filing documentation 
Prior approval supplement with justification ;annual report (long term stability data). 
Handling and Maintenance of Electronic documentation 
Introduction of Electronic documentation: 
An electronic document is any electronic media content (other than computer programs or system files) that 
is intended to be used in either an electronic form or as printed output. 
Originally, any computer data were considered as something internal the final data output was always on 
paper. However, the development of computer networks has made it so that in most cases it is much more 
convenient to distribute electronic documents than printed ones. And the improvements in electronic 
display technologies mean that in most cases it is possible to view documents on screen instead of printing 
them (thus saving paper and the space required to store the printed copies). 
However, using electronic documents for final presentation instead of paper has created the problem of 
multiple incompatible file formats. Even plain text computer files are not free from this problem e.g. under 
MS-DOS, most programs could not work correctly with UNIX-style text files (see newline), and for non- 
English speakers, the different code pages always have been a source of trouble. 
Even more problems are connected with complex file formats of various word processors, spread sheets 
and graphics software. To alleviate the problem, many software companies distribute free file viewers for 
their proprietary file formats (one example is Adobe's Acrobat Reader). The other solution is the 
development of standardized non-proprietary file formats (such as HTML and Open Document), and 
electronic documents for specialized uses have specialized formats–the specialized electronic articles in 
physics use TeX or PostScript. 
GUIDELINES FOR RECORDS OF LONG-TERM OR ENDURING LEGAL VALUE STORED ON 
ELECTRONIC MEDIA 
Planning Process: 
Access: Before you convert your information system, decide how you will provide ready access to the 
records you store off-line. 
Media and systems: Select appropriate media and systems for maintaining your record information. 
Recommended: Magnetic tapes or cartridges are preferred for the long-term storage of electronic records. 
CD-ROMs created with the ANSI 9660 standard can be used as well. The storage capacity of a magnetic 
tape, however, far exceeds that of a CD-ROM. 
Not Recommended: Floppy disks, removable disks and optical disks are not recommended for the long-term 
storage of electronic records. 
Maintenance: 
Access: Maintain your records in a useable format and keep up to date all materials needed to access them, 
including indexes and other documentation, until they are scheduled for disposal or for transfer to the State 
Archives. 
Backups: Maintain in an off-site location backup and archived copies of records and all materials required 
to access them. 
Labeling: Develop procedures for labeling tapes and cartridges. Each external label should carry 
information unique to the tape or cartridge it identifies. At minimum, it should display the name of the 
organizational unit responsible for the data, the system title, the file title, the destruction date or permanent 
status of the record, and its security classification, if applicable.
Documentation
Documentation
Documentation
Documentation
Documentation
Documentation

Contenu connexe

Tendances

Pharmaceutical inspection convention
Pharmaceutical inspection conventionPharmaceutical inspection convention
Pharmaceutical inspection conventionRAGHAV DOGRA
 
Software Evaluation Checklist
Software Evaluation ChecklistSoftware Evaluation Checklist
Software Evaluation ChecklistSalina Saharudin
 
Good documentation practice
Good documentation practiceGood documentation practice
Good documentation practicePharmaceutical
 
Pharmaceutical documentation
Pharmaceutical documentationPharmaceutical documentation
Pharmaceutical documentationK V NANDA KUMAR
 
Good Automated Laboratory Practices
Good Automated Laboratory PracticesGood Automated Laboratory Practices
Good Automated Laboratory PracticesSwapnil Fernandes
 
batch formula record and master formula record
batch formula record and master formula recordbatch formula record and master formula record
batch formula record and master formula recordRashmitaPoojary1
 
Data integrity & ALCOA+
Data integrity & ALCOA+Data integrity & ALCOA+
Data integrity & ALCOA+TuhinReza5
 
Good Documentation Practice
Good Documentation PracticeGood Documentation Practice
Good Documentation PracticeDr. Amsavel A
 
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...ICHAPPS
 
Data Integrity - the ALCOA model
Data Integrity - the ALCOA modelData Integrity - the ALCOA model
Data Integrity - the ALCOA modelpi
 
Audits in pharma industries
Audits in pharma industriesAudits in pharma industries
Audits in pharma industriesNikita Amane
 
BMR (Batch Manufacturing Record)
BMR (Batch Manufacturing Record)BMR (Batch Manufacturing Record)
BMR (Batch Manufacturing Record)Ram Mohan S R
 
SOP- Standard Operation Procedure.
SOP- Standard Operation Procedure.SOP- Standard Operation Procedure.
SOP- Standard Operation Procedure.Arshad Khan
 

Tendances (20)

ALCOA & ALCOA+
ALCOA & ALCOA+ALCOA & ALCOA+
ALCOA & ALCOA+
 
Pharmaceutical inspection convention
Pharmaceutical inspection conventionPharmaceutical inspection convention
Pharmaceutical inspection convention
 
21 CFR PART 11
21 CFR PART 1121 CFR PART 11
21 CFR PART 11
 
Software Evaluation Checklist
Software Evaluation ChecklistSoftware Evaluation Checklist
Software Evaluation Checklist
 
Docmentation complete
Docmentation completeDocmentation complete
Docmentation complete
 
Good documentation practice
Good documentation practiceGood documentation practice
Good documentation practice
 
Sop
SopSop
Sop
 
Pharmaceutical documentation
Pharmaceutical documentationPharmaceutical documentation
Pharmaceutical documentation
 
Good Automated Laboratory Practices
Good Automated Laboratory PracticesGood Automated Laboratory Practices
Good Automated Laboratory Practices
 
batch formula record and master formula record
batch formula record and master formula recordbatch formula record and master formula record
batch formula record and master formula record
 
Data integrity & ALCOA+
Data integrity & ALCOA+Data integrity & ALCOA+
Data integrity & ALCOA+
 
Good Documentation Practice
Good Documentation PracticeGood Documentation Practice
Good Documentation Practice
 
21 code of federal regulation
21 code of federal regulation21 code of federal regulation
21 code of federal regulation
 
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...
21 CFR-FOOD AND DRUG ADMINISTRATION, DEPARTMENT OF HEALTH AND HUMAN SERVICES...
 
Data Integrity - the ALCOA model
Data Integrity - the ALCOA modelData Integrity - the ALCOA model
Data Integrity - the ALCOA model
 
Audits in pharma industries
Audits in pharma industriesAudits in pharma industries
Audits in pharma industries
 
BMR (Batch Manufacturing Record)
BMR (Batch Manufacturing Record)BMR (Batch Manufacturing Record)
BMR (Batch Manufacturing Record)
 
SOP- Standard Operation Procedure.
SOP- Standard Operation Procedure.SOP- Standard Operation Procedure.
SOP- Standard Operation Procedure.
 
Audit
AuditAudit
Audit
 
Handling of Out of Specification Results
Handling of Out of Specification ResultsHandling of Out of Specification Results
Handling of Out of Specification Results
 

Similaire à Documentation

Documentation relating to product development,sop's,cleaning methods,quality ...
Documentation relating to product development,sop's,cleaning methods,quality ...Documentation relating to product development,sop's,cleaning methods,quality ...
Documentation relating to product development,sop's,cleaning methods,quality ...swrk
 
Documentation assignment
Documentation assignmentDocumentation assignment
Documentation assignmentAtul Chaudhary
 
Good documentation practices
Good documentation practicesGood documentation practices
Good documentation practicesPiyush Satvara
 
Writing and enforcing effective so ps
Writing and enforcing effective so psWriting and enforcing effective so ps
Writing and enforcing effective so psGRCTS
 
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...Namdeo Shinde
 
Quality & compliance excellence in pharmaceuticals
Quality & compliance excellence in pharmaceuticalsQuality & compliance excellence in pharmaceuticals
Quality & compliance excellence in pharmaceuticalsAnvita Bharati
 
Product Complaints: Complaint Handling from Intake to Closure
Product Complaints: Complaint Handling from Intake to ClosureProduct Complaints: Complaint Handling from Intake to Closure
Product Complaints: Complaint Handling from Intake to ClosureGRCTS
 
Pharmaceutical documentation
Pharmaceutical documentationPharmaceutical documentation
Pharmaceutical documentationNitinPadole2
 
Document Maintenance in Pharmaceutical Industry
Document Maintenance in Pharmaceutical IndustryDocument Maintenance in Pharmaceutical Industry
Document Maintenance in Pharmaceutical IndustryNAKUL DHORE
 
Amit_Kumar_15052015_AG
Amit_Kumar_15052015_AGAmit_Kumar_15052015_AG
Amit_Kumar_15052015_AGAmit Kumar
 
Regulatory affairs by sachin gundecha
Regulatory affairs by sachin gundechaRegulatory affairs by sachin gundecha
Regulatory affairs by sachin gundechaSachin Gundecha
 
good manufacturing practices presentation
good manufacturing practices presentationgood manufacturing practices presentation
good manufacturing practices presentationUltratech4
 
Importance of documentation for gmp compliance
Importance of documentation for gmp complianceImportance of documentation for gmp compliance
Importance of documentation for gmp complianceJRamniwas
 

Similaire à Documentation (20)

Documentation relating to product development,sop's,cleaning methods,quality ...
Documentation relating to product development,sop's,cleaning methods,quality ...Documentation relating to product development,sop's,cleaning methods,quality ...
Documentation relating to product development,sop's,cleaning methods,quality ...
 
Documentation assignment
Documentation assignmentDocumentation assignment
Documentation assignment
 
Doc in pharma
Doc in pharmaDoc in pharma
Doc in pharma
 
Good documentation practices
Good documentation practicesGood documentation practices
Good documentation practices
 
Documentation
DocumentationDocumentation
Documentation
 
Writing and enforcing effective so ps
Writing and enforcing effective so psWriting and enforcing effective so ps
Writing and enforcing effective so ps
 
Documentation and records
Documentation and recordsDocumentation and records
Documentation and records
 
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...
Practice school session on "GOOD Documentation Practices" dated 08.01.2021 by...
 
Quality & compliance excellence in pharmaceuticals
Quality & compliance excellence in pharmaceuticalsQuality & compliance excellence in pharmaceuticals
Quality & compliance excellence in pharmaceuticals
 
GMP documentation
GMP documentationGMP documentation
GMP documentation
 
Product Complaints: Complaint Handling from Intake to Closure
Product Complaints: Complaint Handling from Intake to ClosureProduct Complaints: Complaint Handling from Intake to Closure
Product Complaints: Complaint Handling from Intake to Closure
 
Pharmaceutical documentation
Pharmaceutical documentationPharmaceutical documentation
Pharmaceutical documentation
 
Document Maintenance in Pharmaceutical Industry
Document Maintenance in Pharmaceutical IndustryDocument Maintenance in Pharmaceutical Industry
Document Maintenance in Pharmaceutical Industry
 
Amit_Kumar_15052015_AG
Amit_Kumar_15052015_AGAmit_Kumar_15052015_AG
Amit_Kumar_15052015_AG
 
Regulatory affairs by sachin gundecha
Regulatory affairs by sachin gundechaRegulatory affairs by sachin gundecha
Regulatory affairs by sachin gundecha
 
Documentation
Documentation Documentation
Documentation
 
Regulatory affairs
Regulatory affairsRegulatory affairs
Regulatory affairs
 
good manufacturing practices presentation
good manufacturing practices presentationgood manufacturing practices presentation
good manufacturing practices presentation
 
Các yêu cầu về tài liệu và hồ sơ các GMP
Các yêu cầu về tài liệu và hồ sơ các GMPCác yêu cầu về tài liệu và hồ sơ các GMP
Các yêu cầu về tài liệu và hồ sơ các GMP
 
Importance of documentation for gmp compliance
Importance of documentation for gmp complianceImportance of documentation for gmp compliance
Importance of documentation for gmp compliance
 

Plus de Malla Reddy College of Pharmacy (20)

Rna secondary structure prediction
Rna secondary structure predictionRna secondary structure prediction
Rna secondary structure prediction
 
Proteomics
ProteomicsProteomics
Proteomics
 
Proteins basics
Proteins basicsProteins basics
Proteins basics
 
Protein structure classification
Protein structure classificationProtein structure classification
Protein structure classification
 
Protein identication characterization
Protein identication characterizationProtein identication characterization
Protein identication characterization
 
Protein modeling
Protein modelingProtein modeling
Protein modeling
 
Primerdesign
PrimerdesignPrimerdesign
Primerdesign
 
Phylogenetic studies
Phylogenetic studiesPhylogenetic studies
Phylogenetic studies
 
Multiple sequence alignment
Multiple sequence alignmentMultiple sequence alignment
Multiple sequence alignment
 
Homology modeling tools
Homology modeling toolsHomology modeling tools
Homology modeling tools
 
Homology modeling
Homology modelingHomology modeling
Homology modeling
 
Genome assembly
Genome assemblyGenome assembly
Genome assembly
 
Genome analysis2
Genome analysis2Genome analysis2
Genome analysis2
 
Genome analysis
Genome analysisGenome analysis
Genome analysis
 
Fasta
FastaFasta
Fasta
 
Drug design intro
Drug design introDrug design intro
Drug design intro
 
Drug design
Drug designDrug design
Drug design
 
Data retrieval
Data retrievalData retrieval
Data retrieval
 
Blast
BlastBlast
Blast
 
Biological databases
Biological databasesBiological databases
Biological databases
 

Documentation

  • 1. UNIT 5: Documetation Documentation is a process that involves the systematic interaction of people, events and documents to create the records of the organization / corporation. It is a collection of document (a piece of written or printed matter) relating to a piece (or) events. Documentation is an important aspect in pharmaceutical industry irrespective of the fact the manufacturing plant is involved in production of dosage form (or) active ingredient. Manufacturer of a drug, ultimately needs quality of his product (or) finished goods. Therefore a good documentation is essential part of a quality control system, hence every manufacturer of drugs should have good documentation. Its aim is to define the specifications for all materials, method of manufacture and control, to ensure that all personnel concerned with manufacture know the information necessary to decide whether or not to release a batch or a drug for sale and-to provide an audit trail that shall permit investigation of the history of any suspected defective batch. DOCUMENT: As discrete packages of recorded information are the tools of quality assurance. Used to establish standard specification and processes that assures compliance with standard specifications and processes. Quality cannot be assured in a regulated industry without good documents and good documentation practices. A document system is an interdependent, interrelated set of documents, each with a defined purpose and a consistent format. OBJECTIVES OF DOCUMENTATION: · To define all materials specifications. · To define manufacture and control methods. · To ensure all personal of manufacture and control department know what to do, and when to do. · To ensure that personal authorized to release a batch for sale (or) reject, it have all information to take decision. · To provide information during investigation, if a batch is suspected to have defect(s). ESSENTIAL CHARACTERISTICS: To design an effective & efficient documentation process first identify the important characteristics of processing event. · The document directing the manufacturing event fulfills current regulatory commitments to the agency; it is appropriately written, reviewed and approved. · The directive document is appropriate for the task to be performed. · The data is authentic; the individual responsible for performing the work has entered the data on an appropriate data collection document. · The data is accurate. · The data is complete. There is no missing information, and there is no work as yet uncompleted that will impact the occurrence of the data presented. · The data is legible, consistently recorded and trustworthy. · The data collected fulfills expectations (specifications). · The data is accessible to those who need to review it, audit it or use it to perform trending analysis. · The original data and the original documents (manufacturing record) is retrievable for review or audit.
  • 2. · The original manufacturing record is secure. Proper control of documentation also requires that responsibility for each processing step to be assigned. “All documentation should be legible, clean, readily identifiable, retrievable and maintained in facilities that provide a suitable environment to minimize deterioration or damage and to prevent loss”. WHY DOCUMENTATION: · There is a saying in the pharmaceutical industry: 'if it hasn't been documented, then it hasn't happened!' · Good documentation practice constitutes an essential part of the QA system. · Documentation system must be proactive vehicle of communication. “YOUR DOCUMENTATION IS AN ADVERTISEMENT FOR YOUR WORKˮ. WHERE DOCUMENTATION ? · Documentation during Project Design. · Documentation during Construction Phase. · Documentation during Commissioning and start-up. · Documentation during Qualification and Validations. · Documentation during Commercial Production. · Documentation during Testing and Release. · Documentation for Regulatory submissions. TYPES OF DOCUMENTS: There are three types of Documents: · Commitment documents. · Directive documents. · Record documents. · Commitment Documents : Relationship between industry and the regulatory authorities. EX: New Drug Applications (NDAs), Drug Master Files (DMFs) etc. · Directive Documents : Relationship between the Management and Employees. EX: Specifications, STPs, Standard Operating Procedures (SOPs), Medicinal Product Records (MPRs) etc. · Record Documents : Relationship between the Employees and the Work they perform. EX: Protocols, Batch Production Records (BPRs), Log Books, Calibration Records etc. · Commitment Documents: · These presents corporate goals, expectations and standard of practice. · It describes what to do. · Consensus of purpose, direction and authorization for projects. · It organize the work in a manner that assures efficient and effective work flow. · Commitments documents can also be written to establish internal commitments. · These documents such as master plans; organize and prioritize the work in a manner that supports good business practice as well as regulatory compliance. · Written to lead and guide the work and workers. · To be used as active documents that are consulted and followed routinely.
  • 3. · Content must be consistent and rigorous in order to lead and guide the work and the workers. · Must describe the work as it will be done. · Deviations from regulatory submissions can have signification impact on product quality. · Any deviation must be documented and managed. · New Drug Applications (NDAs): The New Drug Application (NDA) is the vehicle in the United States through which drug sponsors formally propose that the FDA approve a new pharmaceutical for sale and marketing. The goals of the NDA are to provide enough information to permit FDA reviewers to establish the following: · Is the drug safe and effective in its proposed use(s) when used as directed, and do the benefits of the drug outweigh the 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? · Drug Master Files (DMFs): Drug Master File or DMF is a document prepared by a pharmaceutical manufacturer and submitted solely at its discretion to the appropriate regulatory authority in the intended drug market. There is no regulatory requirement to file a DMF. However, the document provides the regulatory authority with confidential, detailed information about facilities, processes, or articles used in the manufacturing, processing, packaging, and storing of one or more human drugs. Drug Master File (DMF) is a document containing complete information on an Active Pharmaceutical Ingredient (API) or finished drug dosage form. It is known as European Drug Master File (EDMF) or Active Substance Master File (ASMF) and US-Drug Master file (US-DMF) in Europe and United States respectively. The DMF contains factual and complete information on a drug product's chemistry, manufacture, stability, purity, impurity profile, packaging, and the cGMP status of any human drug product. · Directive Document: · Working documents that establish the standards for resources, processing, products & quality system. · Describe how to do it……??? · Describe how to do routine work. · Several types of directive documents. · The different types are determined by the specific, functional purpose of the document in the document system. · In order to facilitate the development, production, testing and distribution of a product in a defined manner. · Reviewed and approved by both management and the individuals responsible for performing the work. · Specifications: A document specification contains several parts: a description of the audience(s) for the document, a detailed outline giving the structure and contents of the document, and a work plan showing who is responsible for each part of the document and what the deadlines are for completing each task. For large documents, there may other managerial information, such as number of pages allocated for each section, graphics budgets, printing costs, and so forth. We do not expect you to produce this level of detail in your document specification. There are three purposes for document specifications:
  • 4. In the workplace, formal document specifications serve three important functions: economy of effort, work planning, and writing organization. · Economy of effort: First, document specifications are used to help you reduce the amount you have to write. When you are requested to write a major report, quite often you will be told something like, ‟Do a draft and then come see me with it”. After talking with your boss, you will have to revise the draft completely so that it will have in it what the boss really wants, since he/she hadn't thought about the project carefully until reading your draft. You can often save yourself (and your boss) much work if you write a detailed document specification instead. A document specification is much easier to create, change, revise, and add to than a draft is. · Work planning: The second major function of a document specification is work planning. This can mean either budgeting your own time, or, in large formal reports, distributing the work among many people. Multi-author writing is probably the most common form of workplace writing. The more you know about the specification, the more likely you are to get only your fair share of the writing and no more. For the purposes of the class, the document specification will serve to distribute the work between you and your partner. You should bear three things in mind in dividing the work: even work load, respective areas of expertise, and getting things done in the order that you need them. These points sound obvious, but achieving a fair distribution of the work is not always easy. It involves keeping to a schedule, for one thing, and staying in close touch with your partner, for another. · Writing Organization: The third major function is the organization of the report itself. · Standard Operating Procedures (SOPs): Standard Operating Procedures (SOPs) are issued to specifically instruct employees in areas of responsibility, work instructions, appropriate specifications and required records A Standard Operating Procedure (SOP) is a set of written instructions that document a routine or repetitive activity followed by an organization. The development and use of SOPs are an integral part of a successful quality system as it provides individuals with the information to perform a job properly, and facilitates consistency in the quality and integrity of a product or end-result. The term “SOP” may not always be appropriate and terms such as protocols, instructions, worksheets, and laboratory operating procedures may also be used. For this document “SOP” will be used. SOPs describe both technical and fundamental programmatic operational elements of an organization that would be managed under a work plan or a Quality Assurance (QA) Project Plan [EPA Requirements for QA Project Plans (QA/R-5) (EPA 2001a)], or Chapter 5 of the EPA Quality Manual for Environmental Programs, (EPA Manual 5360 A) and under an organization’s Quality Management Plan [EPA Requirements for Quality Management Plans (QA/R-2) (EPA 2001b)], or Chapter 3 of the EPA Quality Manual. This document is designed to provide guidance in the preparation and use of an SOP within a quality system. Purpose: SOPs detail the regularly recurring work processes that are to be conducted or followed within an organization. They document the way activities are to be performed to facilitate consistent conformance to technical and quality system requirements and to support data quality. They may describe, for example, fundamental programmatic actions and technical actions such as analytical processes, and processes for maintaining, calibrating, and using equipment. SOPs are intended to be specific to the organization or facility whose activities are described and assist that organization to maintain their quality control and quality assurance processes and ensure compliance with governmental regulations. If not written correctly, SOPs are of limited value. In addition, the best written SOPs will fail if they are not followed. Therefore, the use of SOPs needs to be reviewed and re-enforced by management, preferably the direct supervisor. Current copies of the SOPs also need to be readily accessible for reference in the work
  • 5. areas of those individuals actually performing the activity, either in hard copy or electronic format, otherwise SOPs serve little purpose. · Record document: Protocol: Protocols are written records clearly defining the objectives and methods that will be used for the validation programs. An important part of the protocol is the description of the testing method including who will test the system, how they will test it and what data is to be collected and reported. Computerized system protocols often include the three distinct stages as described in PMA reports: Installation Qualification (IQ), Operational Qualification (OQ),and Performance Qualification (PQ). Protocol Changes are documented requirements specifying who and how changes to parameters, thresholds, and acceptance criteria are made after approval. It is not impossible to make changes after or during testing, but these changes must be properly implemented and approved to be validatable. What Good Documentation requires: APPROVAL - this applies particularly to work instructions, procedures, manufacturing formulae and specifications. Approval should be by the relevant technical, management and quality personnel, to ensure that documents comply with the principles of GMP and the specific product marketing and manufacturing authorizations. CLARITY - they should not be open to misinterpretation by the users. They should be written in a way that makes them easy to check, particularly when they will form part of a product manufacturing history. Good documentation design will help to minimize errors. REGULAR REVIEW AND UPDATE - documents must be kept up-to-date with changes in regulations or processes and should be distributed in a controlled manner to ensure that only the most recent versions are available for use. They must also be available to those who need them, where they need them! FORMAL PRESENTATION - controlled documents should be prepared in accordance with a written procedure, now a days probably using a computerised documentation control system. Records should be made at the time of each action - do not rely on memory for their completion. Records relating to manufacturing or testing operations should be kept for at least one year after their expiry. If documents or data are stored electronically, the computer system must be validated to assure data security and integrity. Provisions must also be made to retrieve the stored data, possibly years after they have been generated. PROTOCOL FOR DOCUMENTATION: Protocol is a detailed plan and instructions to the employees working in pharmaceutical industry. It is a guidebook for those involved in work. The manufacturing records relating to manufacture of sterile products shall indicate the following details:- (1) Serial number of the Batch Manufacturing Record. (2) Name of the product. (3) Reference to Master Formula Record. (4) Batch/Lot number. (5) Batch/Lot size. (6) Date of commencement of manufacture and date of completion of manufacture. (7) Date of manufacture and assigned date of expiry. (8) Date of each step in manufacturing. (9) Names of all ingredients with the grade given by the quality control department. (10) Quality of all ingredients. (11) Control reference numbers for all ingredients. (12) Time and duration of blending, mixing, etc. whenever applicable. (13) pH of solution whenever applicable. (14) Filter integrity testing records. (15) Temperature and humidity records whenever applicable.
  • 6. (16) Records of plate-counts whenever applicable. (17) Results of pyrogen and/or bacterial endotoxin & toxicity. (18) Results of weight or volume of drug filled in containers. (19) Bulk sterility in case of aseptically filled products. (20) Leak test records. (21) Inspection records. (22) Sterilization records including autoclave leakage test records, load details, date, duration, temperature, pressure, etc. (23) Container washing records. (24) Total number of containers filled. (25) Total numbers of containers rejected at each stage. (26) Theoretical yield, permissible yield, actual yield and variation thereof. (27) Clarification for variation in yield beyond permissible yield. (28) Reference numbers of relevant analytical reports. (29) Details of reprocessing, if any. (30) Name of all operators carrying out different activities. (31) Environmental monitoring records. (32) Specimens of printed packaging materials. (33) Records of destruction of rejected containers printed packaging and testing. (34) Signature of competent technical staff responsible for manufacture and testing. In addition to the routine good manufacturing practices documentation, manufacturing records shall show the following additional information:- (1) Temperature and humidity in the manufacturing area. (2) Periodic filled weights of the formulation. (3) Records of rejections during on line check weighing. (4) Records of rejection during spray testing. REFERENCES: · www.s uperiorcontrols.com/news.php?id=31 & page=4 · http://www.medicalnewstoday.com/articles/172522.php · http://www.drugs.com/nda/acetavance_090715.html · http://www.sciencemag.org/cgi/content/full/326/5951/370 · http://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/u cm122886.htm Formulation development for global fillings THE IMPACT OF WORLD REGULATIONS TO DEVELOP AND LAUNCH HARMONIZED COSMETIC PRODUCTS Business Strategy · Since long time ago, we have been challenged: “ How can we develop a cosmetic product that can have its inventory ready to be exchanged around the globe.” · Same package with all information in · Same formula “In other words: How to make one fit all” Understand what is required in each country to sell cosmetics Global Cosmetic Regulation Scenario We have a lack of global harmonization on Cosmetics: · USA
  • 7. · Europe · Asia · Latin America Robust R&D system is required Latin America · Mercosur · CAN · Central America · Mexico · Chile · Panama · All others Mercosur · Argentina, Brazil, Paraguay and Uruguay · PreMarketing approval process · Lists of ingredients harmonized · In place process to update the list · Unique requirements still present in each country · Administrative procedures to get product registration are not harmonized · Requirements change country by country · Labeling requirements not harmonized · Claims /Safety support data requirements not harmonized CAN · Bolivia, Colombia, Ecuador, Peru and Venezuela* · PreMarketing Approval process · Recognition Process in place · Adopts List of Ingredients from Europe and USA · Administrative procedures to get product registration are almost quite harmonized: · Requirements quite clear · Labeling requirements almost quite harmonized · Claims /Safety support data requirements almost quite harmonized Central America (RTCA) · Costa Rica, El Salvador, Guatemala, Honduras, Nicaragua. · New Harmonized cosmetic regulation since early 2009 · PreMarketing approval process · Adopts List of Ingredients from Europe and USA · Administrative procedures to get product registration are not harmonized: · Requirements change country by country · Labeling not harmonized · Claims /Safety support data requirements not harmonized Mexico · Pos-Marketing Surveillance process for cosmetics · Pre-Marketing approval process for Hygienic
  • 8. · Has list of allowed, restricted and not allowed ingredients updated on March 21, 2007 · Specific labeling requirements ( INCI translated to Spanish) Chile and Panama · PreMarketing approval process · Adopts List of Ingredients from Europe and USA · Specific labeling requirements R&D Process · Product Development · R&D systems should be able to provide to the Formulator the ingredient’s regulation around the global/region so that only approved ones can be used · Product Safety: · Make sure that ingredients that are not regulated are safe. · Make sure that final formulation is safe · Make sure claims are substantiated Regulatory Affairs · Make sure to update database system · Ingredients update · Safety test requirements · Specific country/region product registration requirements: · Words/expression restriction / forbidden · Claims requirements · Unique requirements
  • 11. Harmonized Product · Last but not least · Define Manufacturing strategy · This will drive the last regulatory requirements to have harmonized product: · Same package · Same formula · Same label copy · Approved in the countries according strategy The impact of world regulation to develop and launch harmonized cosmetic products · Time to launch harmonized product is too long impacting: · Marketing strategy · Supply Chain strategy · Adding Premium costs · Service issues Patents are exclusive property rights in intangible creations of the human mind. They exist only as provided in the laws of sovereign states, and can be enforced only to the extent that application has been made and a patent granted covering the territory of an individual state. Patent rights are limited in duration, with the global standard being 20 years from the date of application. The new product, article of manufacture or process described in the patent application must be something that has never been previously disclosed anywhere in the world and something that would not be obvious to a person ordinarily skilled in the field involved. Determinations of whether these requirements have been met are made by comparing the claims
  • 12. of the patent applicant against the body of published literature in the field, including previously issued patents. This process is called examination, and it assures that no one is able to claim patent rights on anything that already is existence. Patents work differently indifferent industries. In the electronic industry patents are often shared among competitors through pooling or cross licensing. This sharing is necessary because a given product often contains many patented technologies. However, in the pharmaceutical, chemical and biotechnology industries the patent normally equals the product, and protects the extensive investment in research and clinical testing required before placing it on the market. Patent protection for chemical and pharmaceutical products is especially important compared with other industries because the actual manufacturing process is often easy to replicate and can be copied with a fraction of the investment of that required for the research and clinical testing. The extensive cost required to produce a new pharmaceutical product has meant that private sector investment in pharmaceutical innovation has been disproportionately directed to products meeting the needs of patients in developed countries, particularly in the United States, which combines strong patent protection with a market free of price controls. Until the TRIPS Agreement in 1994 many developing countries provided no patent protection for pharmaceutical products. And, while countries that have joined the WTO have obligated themselves to provide such protection, least developed countries are not required to meet this obligation until 2016. The continuing lack of patent protection for pharmaceutical products makes it very difficult to establish research-based industries in most developing countries. Most medical research in these countries takes place in the public sector. The lack of any means of patenting these inventions and the related lack of experience in licensing them to the private sector, suppresses the development of commercial enterprises focused on alleviating the disease burdens common to developing countries. The controversy over availability of patented therapies for the treatment of HIV disease has resulted renewed interest in the compulsory licensing of pharmaceutical products. After two years of discussion, the WTO Council recently affirmed that the TRIPS Agreement permits such compulsory licenses in health emergencies, even in cases where the compulsory license is for an imported product. However, to date, no compulsory licenses actually have been issued, even though the threat of compulsory licensing has been used as a means of seeking lower prices. One danger in compulsory licensing is that it will discourage further the commercial R & D necessary to new drugs to fight global epidemics. Another danger is that compulsory licensing can be used to seek price levels below what a given national market is capable of supporting, further concentrating the burden of financing pharmaceutical innovation on developed country consumers and discouraging development of drugs targeted at the disease burdens of countries using compulsory licenses. There are promising developments in countries such as India and Brazil that are beginning to use patents to develop commercial pharmaceutical industries that produce products directed at local diseases and available at price that patients in those countries can afford. Foundations and nonprofit organizations such as the Bill and Melinda Gates Foundation and One World Health, Inc. are supporting such efforts. These efforts show that developing countries have the capacity to build research-intensive pharmaceutical industries capable of operating profitably in the conditions of the local market. However, for such local industries to take root and grow, effective patent protection must be made available, the commercialization of publicly funded research must be encouraged, and compulsory licensing must be kept to a minimum. Wealthy countries can assist this process by subsidizing local markets for the purchase of drugs through the Global Fund, and by direct programs of assistance such as that recently proposed by President Bush. Consumers in all countries can share the burden of drug development equitably by paying for medicine at a price level consistent with their means, rather than attempting to shift the costs of drug development to others. REFERENCES: 1. http://www.personalcarecouncil.org
  • 13. 2. http://ec.europa.eu/enterprise/cosmetics/ 3. http://www.cirsafety . org/ NDA INTRODUCTION: New Drug Application is a Critical component for drug approval process which required to submit to USFDA before drug commercialization. The data gathered during the animal studies and human clinical trials of an Investigational New Drug (IND) become part of the NDA. The regulation and control of new drugs in the United States has been based on the New Drug Application (NDA). Since 1938, every new drug has been the subject of an approved NDA before U.S. commercialization. Goals of the NDA: To provide enough information to permit FDA reviewer to reach the following key decisions: · Whether the drug is safe and effective in its proposed use(s), and whether the benefits of the drug outweigh the risks. · Whether the drugs proposed labeling (package insert) is appropriate, and what it should contain. · Whether the methods used in manufacturing the drug and the controls used to maintain the drug's quality are adequate to preserve the drug's identity, strength, quality, and purity. NDA Classifications: · New Molecular Entity · New Salt of Previously Approved Drug (not a new molecular entity) · New Formulation of Previously Approved Drug (not a new salt OR a new molecular entity) · New Combination of Two or More Drugs Already Marketed Drug Product - Duplication (i.e., new manufacturer) · New Indication (claim) for Already Marketed Drug (includes switch in marketing status from prescription to OTC) · Already Marketed Drug Product - No Previously Approved NDA. Assembling Applications for Submission: Assembling Applications for Submission the FDA requires drug sponsors to submit multiple copies of the NDA The archival copy, the review copy, the field copy. In 1997 the FDA Center for Drug Evaluation and Research (CDER) published guidelines that allow sponsors to submit NDAs electronically instead of on paper. The Archival Copy: The Archival Copy Contains all sections of the NDA, including the cover letter, Form FDA-356h (Application to Market a New Drug, Biologic, or an Antibiotic for Human Use), the administrative sections, Comprehensive NDA index, and All technical sections. It must contain four copies of the Labeling section. It must contain three additional copies of the CMC and Methods Validation Package in a separate binder. The archival copy is the only copy that contains the Case Report Tabulation and Case Report Forms. The Review Copy: The Review Copy Intended for reviewers in the corresponding technical disciplines. In addition to the appropriate technical section, each review copy also includes The cover letter, Form FDA-356h, The administrative sections, Comprehensive NDA index Individual table of contents, The Labeling section, and The Application Summary. The Field Copy:
  • 14. The Field Copy required since 1993 for use by FDA inspectors during pre approval facilities inspections. It includes the Cover letter and Form FDA-356h, the administrative sections, the comprehensive NDA index Individual table of contents, The Labeling section, The Application Summary, and CMC and Methods Validation Package. NDA CONTENTS: The NDA may have as many as 20 different sections in addition to the Form FDA-356h itself. The specific contents of the NDA will depend on the Nature of the drug and the information available at the time of submission. The application Form FDA-356h serves as Checklist as well as Certification that the sponsor agrees to comply with a range of legal and regulatory requirements. Section 1: Index Section 2: Labeling Section 3: Application Summary Section 4: Chemistry, Manufacturing, and Controls (CMC) Section 5: Nonclinical Pharmacology and Toxicology Section 6: Human Pharmacokinetics and Bioavailability Section 7: Microbiology Section 8: Clinical Data Section 9: Safety Update Reports Section 10: Statistics Section 11: Case Report Form Tabulations Section 12: Case Report Forms (CRFs) Section 13: Patent Information Section 14: Patent Certification Section 15: Establishment Description Section 16: Debarment certificate Section 17: Field copy certification Section 18: User fee coversheet Section 19: Financial Disclosure Section 20: Other/ Pediatrics Section 1: Index: The NDA index is a comprehensive table of contents that enables the reviewers to find specific information in this massive document quickly. The NDA index should follow immediately after the Form FDA-356h and the administrative items. It must show the location of every section in the archival NDA by volume and by page number. Index It should guide reviewers the data in the technical sections, the summary, and the supporting documents each separately bound technical section should also contain a copy of the overall NDA index in addition to its own table of contents based on the index. Section 2: Labeling: The labeling section must include all draft labeling that is intended for use on the product container, Cartons or packages, the proposed package insert. Section 3: Application Summary: The application summary is an abbreviated version of the entire application. This overview is one of the few elements of the application that all reviewers receive, and it should give them a clear idea of the drug and its application. The summary usually comprises 50 to 200 pages. The draft product labeling include the following sections 1. Description 2. Clinical Pharmacology 3. Indications and Usage 4. Contraindications 5. Warnings 6. Precautions
  • 15. 7. Adverse Reactions 8. Drug Abuse and Dependence 9. over dosage 10. Dosage and Administration 11. How Supplied (primary and secondary packages) For each section of the labeling, include annotations referring to information in the summary and technical sections of the application that support the inclusion of each statement in the labeling with respect to Animal pharmacology and/or animal toxicology, Clinical studies, Integrated Summary of Safety (ISS) and Integrated Summary of Effectiveness (ISE) Safety (ISS) and Integrated Summary of Effectiveness (ISE) Pharmacologic class Scientific rationale Intended use Potential clinical benefits. Foreign marketing history: The summary must include a list of any countries in which the drug is or was marketed, along with the dates when it was marketed, if they are known. It must also include a list of any countries in which the drug has been withdrawn from marketing for any reason relating to safety or efficacy or in which an application has been rejected. Section 4: Chemistry, Manufacturing, and Controls (CMC): The first technical section of the NDA.It includes information on the composition, Manufacturing, and Specifications of the drug substance and the drug product. The CMC information must include Description of the drug substance or active ingredient, Its stability, Physical and chemical characteristics, Provide the names/designations of the drug substance, including Generic/common name Chemical name (IUPAC/USAN/CAS) Code(s) (CAS/internal).It Provide a structural overview including Molecular structure, Empirical formula, Molecular weight, Elemental composition. The description of the drug substance physical and chemical characteristics should include: Appearance, including color, crystalline form, and odor Melting/boiling point Refractive index, viscosity, and specific gravity Polymorphs, including modifications (forms) and relative kinetic/ thermodynamic stabilities. The physical and chemical characteristics should also include Solubility, Ionization constants, and Partition coefficients at various pHs. Solubility in common organic solvents as well as in various aqueous mediaWater0.1 N HCl 0.02 N HCl SGF without pepsin Water buffered to various acidic/neutral/basic pHs. It Provide a reference standard (RS) to elucidate the drug substance chemical structure, including Preparation method, Test methods, Test results as shown by a certificate of analysis (C.O.A).Provide proof that the reference standard was adequately tested and characterize the spectra completely. Provide structural elucidation using a reference standard as applicable. Measures might include X-ray (in the case of absolute configuration or polymorphism) UV/visible spectrum, FTIR spectrum, 1H NMR/13C NMR spectrum, Low-resolution/high-resolution mass spectrum, Elemental analysis. The CMC information must also include theNames, addresses and functions of each site where the drug substance is manufactured or tested. The description of the drug substance manufacturing methods must include Synthesis scheme, Synthesis description, Typical executed manufacturing record Compilation of and analytical controls for starting materials, Reagents, Solvents Catalysts, and Intermediates Suppliers for starting materials. The discussion of drug substance analytical controls should include the following: Specifications Methods Rationale for methods/specifications Method validations Batch analytical data (including impurity profiles cross-referenced with toxicology studies)Sampling plan Provide information on drug substance stability including: Ambient/accelerated stability data Retest dating Highly stressed (e.g., acid, base, reflux) data Provide a listing of all inactive ingredients. For compendial (e.g., UPS/NF) inactive ingredients, reference the appropriate current compendial monographs and provide more precise specifications as necessary. For noncompendial ingredients that fall under 21 CFR such as D&C and FD&C dyes, reference the appropriate section of 21 CFR and provide any additional specifications beyond the scope of the CFR.For noncompendial items that are not regulated by 21 CFR, provide appropriate analytical specifications and methods. Provide information on the drug product manufacturing methods: Summary and schematics of manufacturing procedure Master batch record for proposed marketed products, including actual operating conditions, type and size of equipment, and in process controls and tests Executed batch record. The section on drug product packaging must include: Summary of container/closure system(s)Listing of packaging
  • 16. components and component/resin suppliers Specifications for each packaging component DMF authorization letters Description of the packaging process Test methods (as appropriate)Developmental data that confirms the suitability of the packaging. This includes water vapor permeation data for plastic containers/ closures and compatibility testing for solutions, suspensions, emulsions, etc. The drug product stability information will differ slightly from the drug substance stability information. Unstressed/stressed stability data Statistical analysis to establish consistency of data Expiration dating Post approval stability commitment/protocol. For an NDA, provide a list of all drug product investigational formulations used in clinical studies, along with the quantitative composition of each formulation. Every NDA must include an environmental assessment (EA)The EA, also called the environmental impact analysis report, includes an analysis of the manufacturing process and ultimate use of the drug product as well as a discussion of how the process and the drug product may affect the environment. Methods validation package: The final component of the CMC technical section is the methods validation package. The package must comprise: Specifications and test methods for each component used in the drug product Specifications and methods for the drug product Validation of test methods Names and addresses of component suppliers Names and addresses of the suppliers of the container closure system. Section 5: Nonclinical Pharmacology and Toxicology: The second technical section of the NDA provides a description of all animal and in vitro studies with the drug. Include a narrative summary of notable findings in all studies and a discussion of notable findings across the various studies. Provide individual study reports,including Pharmacology, Toxicology, and ADME studies. For the pharmacology studies, following data is required 1.Effects related to the therapeutic indication, such as the pharmacodynamic ED 50 in dose-ranging studies and the mechanism of action (if known) 2.Secondary pharmacological actions in order of clinical importance as possible adverse effects or as ancillary therapeutic effects 3. Interactions with other drugs. The Toxicology information must include information on Acute toxicity, Multidose toxicity (including subchronic, chronic, and carcinogenicity) Special toxicity studies, as well as Reproduction studies and mutagenicity studies. Present toxicology data by intended route of administration in the following order: 1. Oral 2.Intravenous 3.Intramuscular 4.Interperitoneal 5.Subcutaneous 6.Inhalation 7 Topical 8 Other in vivo 9.In vitro. Example: For acute toxicity studies, present the animal study data in the following order: 1. Mouse 2.Rat 3.Hamster 4.Other rodent(s) 5.Rabbit 6.Dog 7.Monkey 8.Other nonrodent mammal(s) 9 Non mammals. Section 6: Human Pharmacokinetics and Bioavailability : This technical section includes data from Phase I safety and tolerance studies in healthy volunteers and ADME studies. It should include a table of PK parameters, giving the values for the major parameters (mean and % cv) such as Peak concentration (Cmax) Area under the curve (AUC) Time to reach peak concentration (tmax) Elimination constant (Ke) Distribution volume (Vd) Plasma and renal clearance Urinary excretion. Drug formulation information should include a list of all formulations used in clinical trials and in in vivo bioavailability and PK studies. The analytical methods used must be summarized in each in vivo biopharmaceutic study. Include detailed information, such as Sensitivity Linearity Specificity and Reproducibility of the analytical test methods used in each study. Provide dissolution data on each strength and dosage form for which an approval is sought. Include a comparative dissolution study with the lot in the in vivo biopharmaceutic study. Include summary of the product Dissolution performance Dissolution method and Dissolution specifications. This technical section must include individual study reports from five types of biopharmaceutic. Pilot or background studies Bioavailability/bioequivalence Pharmacokinetic studies Other in vivo studies In vitro studies . Section 7: Microbiology :
  • 17. Required for anti infective drug products.Antimicrobial drugs differ from other classes of drugs in that they are designed to affect microbial physiology rather than patient physiology. In vitro and in vivo studies are critical in establishing the new drug effectiveness, especially if the microorganism has the potential to develop, or has developed, resistance to other antimicrobial drugs. This section requires the following technical information and data: 1. A complete description of the biochemical basis of the drug action on microbial physiology. 2. The drug antimicrobial spectrum. Include results of in vitro studies demonstrating the concentrations of the drug that are required for effective use. 3. Describe any known mechanisms of resistance to the drug and provide information or data of any known epidemiologic studies demonstrating prevalence to resistance factors. 4. Clinical microbiology laboratory methods, such as in vitro sensitivity discs, necessary to evaluate effective use of the drug. Section 8: Clinical Data: This technical section of the NDA comprises ten elements. The document largest and most complex section. List of investigators and list of INDs and NDAs Background/overview of clinical investigations Clinical pharmacology section Controlled clinical trials Uncontrolled clinical trials Other studies and information section Integrated summary of effectiveness data Integrated summary of safety information Drug abuse and over dosage information Integrated summary of benefits and risks of the drug List of investigators and list of INDs and NDAs-- The list of investigators should include all investigators who have used any dosage form. Alphabetize the list and note each investigator address, the type of study, the study identifier, and its location in the NDA .Provide a list of all known INDs under which the drug, in any dosage form, has been studied. Background/overview of clinical investigations-- Describe the general approach and rationale used in developing the clinical data. Explain how information about the drug derived from clinical pharmacology studies led to critical features of the clinical studies.Support the basis for the design features of the clinical trials, such as number of patients, duration, selection criteria, and controls Clinical pharmacology-- Clinical pharmacology Should include ADME studies, pharmacodynamic dose range, and dose response studies, and any other studies of the drug action. The format and order of presentation is as follows: 1. Table of all studies grouped by study type. Provide the investigators, study numbers, start date, and location of the report in the NDA. 2. For each group of studies, a brief synopsis of each study 3. An overall summary of the clinical pharmacology data Controlled clinical trials --Controlled clinical trials Provide the following material in the order presented below: 1. A table of all studies 2. Full clinical trial reports of all controlled studies in the following order: Completed studies Ongoing studies with interim results Incomplete or discontinued studies 3. Full reports of dose-comparison concurrent control studies, followed by those for aoeno- treatment concurrent control, active control studies, and historical control studies Uncontrolled clinical trials --Uncontrolled clinical trials They may be used to provide support for controlled studies and to provide critical safety information. This section should include a table of all studies. Group full reports of studies according to completeness and availability of Case Report Forms (CRFs). Other studies and information: Include a description and analysis of any additional information that the applicant has obtained from any source, foreign or domestic, that is relevant to evaluating the products safety and effectiveness. It should include information on commercial marketing experience and foreign regulatory actions, including List of countries in which the drug has been approved Details of any rejected registrations Copies of approved labeling (package inserts) from major regions such as Europe, Canada, Australia, New Zealand, and Japan Any other reports from the literature not provided Section 9: Safety Update Reports : A pending application must be updated when new safety data becomes available that could affect any of the following: Statements in draft labeling Contraindications Warnings Precautions Adverse events. Safety update reports are not to be used to submit any new final reports that may impact FDA review time unless the FDA agrees at the pre-NDA meeting that it will accept the reports in this
  • 18. manner. Safety updates are submitted 4 months (120 days) after the initial application, following the receipt of an approval letter and at any other time that the FDA requests such an update. Section 10: Statistics : This technical section includes descriptions and documentation of the statistical analyses performed to evaluation the controlled clinical trials and other safety information. It must include copies of All controlled clinical trial reports Integrated efficacy and safety summaries Integrated summary of risks and benefits. Section 11: Case Report Form Tabulations : This section must include complete tabulations for each patient from every adequately or well-controlled Phase II and Phase III efficacy study, and from every Phase I clinical pharmacology study. It also must include tabulations of safety data from all clinical studies. Section 12: Case Report Forms (CRFs) : It is necessary to include the complete CRF for each patient who died during a clinical study and for any patients who were dropped from the study . The report must be submitted regardless of whether the AE is considered to be related to the study drug, even if the patient was receiving a placebo or comparative drug. Additional CRFs must be provided at the request of the FDA. several other supporting items as appropriate Item 13: Patent information Item 14: Patent certification Item15: Establishment description Item 16: Debarment certification Item 17: Field copy certification Item 18: User fee cover sheet (Form FDA-3397) Item19: Financial disclosure (Form FDA 3454, form FDA-3455) Item 20:Other/pediatric use. 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 the NDA is found to be insufficiently complete (and reasons for this can vary from a simple administrative mistake in the application to a requirement to reconduct much of the testing), then the FDA rejects the application with the issue of a Refuse to File letter which is sent to the applicant explaining where the application has failed to meet requirements. Assuming that 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. A standard review implies an FDA decision within about 10 months while a priority review should complete within 6 months. The NDA in CTD Format : The NDA in CTD Format ICH has developed a Common Technical Document to streamline regulatory submissions in Europe, the U.S. and Japan. CTD is an information format that contains clinical, nonclinical, and manufacturing technical data.The CTD format features well-defined modules, with a highly specific structure and numbering of sections within the modules. It makes a clear distinction between subjective information sections and objective information sections. Use of the CTD format benefits both regulatory agencies and the pharmaceutical industry. In addition to enhancing reviews, the CTD use of common elements facilitates communications between the agencies and the applicants and simplifies the exchange of information between regulatory authorities. The document also provides a common basis for continuous improvement of Good Regulatory Practices. REFERENCES: Douglas J. Pisano, David S. Manlus “FDA Regulatory Affairs, A guide for Prescription Drugs, Medical Devices and Biologics-New drug Application “Second edition-Marcel Dekker, page no 69-108. http://www.fda.gov/cder/guidance/index.htm. 31
  • 19. Abbreviated New Drug Application (ANDA): Definition : “A drug product that is comparable to a brand/reference listed drug product in dosage form, strength, route of administration, quality and performance characteristics, and intended use” It is termed as "abbreviated" because they generally not required to include preclinical (animal) and clinical (human) data to establish safety and effectiveness. It is an application for generic versions of off-patent drugs to receive FDA approval with less costly or no preclinical and clinical testing. The intention is to minimize duplication of available information. No safety or efficacy trials are performed. Complete chemistry, manufacturing, and controls information and information regarding the bioavailability of solid dosage forms. Basic Generic Drug Requirements are:-- · Same active ingredient(s) · Same route of administration · Same dosage form · Same strength · Same conditions of use · Inactive ingredients already approved in a similar NDA Goal of ANDA: Provide enough information to permit FDA to make the following key decisions: · Whether the drug is safe and effective in its proposed use(s), and whether the benefits of the drug outweigh the risks. · Whether the drug's proposed labeling (package insert) is appropriate, and what it should contain. · Whether the methods used in manufacturing the drug and the controls used to maintain the drug's quality are adequate to preserve the drug's identity, strength, quality, and purity. · To reduce the price of the drug. · To reduce the time development. · Increase the bioavailability of the drug in comparison to references list drug. Abbreviated or Supplemental NDA: · Abbreviated NDA: includes.. · Generic drug · New combination of approved drugs · Proportion of ingredients in combination is changed · Supplemental NDA: includes.. · New intended use of the drug (labeling change) · Dose, method or duration of administration is changed · Change in manufacturing process or location NDA vs. ANDA Review Process: Requirements for NDA: · Labeling · Pharmacology and toxicology · Chemistry · Manufacturing · Controls · Microbiology
  • 20. · Inspection · Testing · Animal studies · Human studies · Bioavailability Requirements for ANDA: · Labeling · Pharmacology and toxicology · Chemistry · Manufacturing · Controls · Microbiology · Inspection · Testing · Bioequivalence NDA ANDA Applicable for new drug Applicable for generic drug Takes longer time ( 12-15 years) Comparatively less time taken(1-2 years) More expenditure of money Comparatively less Cost of drugs are more Cost of drugs are less Nonclinical studies and clinical investigations are essential Nonclinical studies and clinical investigations are nonessential except bioavailability and bioequivalence Format of ANDA: Three copies of application are required, an archival copy, a review copy and a field copy. FDA will maintain guidance documents on the format and content of applications to assist applicants in their preparation. ANDA includes the following: · Application form: the applicant shall submit a completed and signed application form that contains the information. The applicant shall state whether the submission is an abbreviated application or a supplement to an abbreviated application. · Table of contents: the archival copy of the ANDA is required to contain a table of contents that shows the volume number and page number of the contents of the submission. · Basis for ANDA submission: an ANDA must refer to a listed drug. Ordinarily, that listed drug will be the drug product selected by the agency as the reference standard for conducting bioequivalence testing. The application shall contain: · The name of the reference drug, including its dosage from and strength. For an abbreviated new drug application based on the reference listed drug must be the same as the listed drug approved in the petition. · A statement as to whether, according to the information published in the list, the reference listed drug is entitled to a period of marketing exclusivity. · Conditions of use: · A statement that the conditions of use prescribed, recommended, or suggested in the labeling proposed for the drug product have been previously approved for the reference listed drug. · A reference to the applicant’s annotated proposed labeling and to the currently approved labeling for the reference listed drug.
  • 21. · Active ingredients · Route of administration, dosage form and strength. · Bioequivalence · Labeling · Samples: need not be submitted until requested by FDA. · Patent certification · Financial certification or disclosure statement. ANDA contents: Section 1: Overall ANDA index:- The NDA index is a comprehensive table of contents that enables the reviewers to find specific information in this massive document quickly. Section 2: Labeling:- It must include all draft labeling that is intended for use on the product container, cartons or packages, including the proposed package insert. Section 3: Application summary:- Proposed annotated package insert- · Pharmacology class, scientific rational, intended use, and potential clinical benefits · Foreign marketing history · Chemistry, Manufacturing and control summary · Nonclinical pharmacology and toxicology summary · Human pharmacokinetics and bioavailability summary · Microbiology summary · Clinical data summary and results of statistical analysis · Discussion of benefit/risk relationship Section 4: Chemistry, manufacturing and controls:- · Chemistry, manufacturing and control information · Samples · Methods validation package Section 5: Nonclinical pharmacology and toxicology · Provide individual study reports, including pharmacology, toxicology, ADME studies. · Effects related to the therapeutic indication, such as the pharmacodynamic ED50 in dose- ranging studies and the mechanism of act ion (if know n) · Interactions with other drugs (or cross-reference the location of the information in any of the above subsection Section 6: Human Pharmacokinetics and bioavailability:- includes data from Phase I safety and tolerance studies in healthy volunteers. Element in the section tabulated summary of studies showing all in vivo biopharmaceutics studies performed. · Summary of analytical method used in in vivo biopharmaceutic study · Pilot or background studies · Bioavailibility or bioequivalence studies · Pharmacokinetic studies · In vitro studies Section 7: Microbiology:- Includes for anti infective drug products. It requires the following technical information and data:- · A complete description of the biochemical basis of the drug action on microbial physiology · The drugs antimicrobial spectrum
  • 22. · Describe any known mechanism of resistance to the drug and provide information/data of any known epidemiologic studies demonstrating prevalence to resistance factor · Clinical microbiology laboratory methods Section 8: Safety data ·· Statements in draft labeling · Contra indications · Warnings · Precautions · Adverse events Section 9: Statistical data · All controlled clinical trial reports · Integrated efficacy and safety summaries · Integrated summary of risks and benefits Section 10: Case report tabulation:- Includes complete tabulation for each patient from every adequately are well controlled phase II and Phase III efficacy, clinical pharmacology study. It also tabulation of safety data from all clinical studies. Section 11: Case report forms:- Includes the complete CRF for each patient who died during a clinical study or adverse event, regardless of whether the AE is considered to be related to the study drug, even if the patient was receiving a placebo or comparative drug. Bioequivalence: A generic drug is considered to be bioequivalent to the brand name drug if: · The rate and extent of absorption do not show a significant difference from listed drug, or · The extent of absorption does not show a significant difference and any difference in rate is intentional or not medically significant. Bio equivalent Inequivalent Resources for ANDA Submissions:- The following resources have been gathered to provide you with the legal requirements of an ANDA application, assistance from CDER to help you meet those requirements, and internal ANDA review principles, policies and procedures. Guidance Documents for ANDAs:- Guidance documents represent the Agency's current thinking on a particular subject. These documents are prepared for FDA review staff and applicants/sponsors to provide guidelines to the processing, content, and evaluation/approval of applications and also to the design, production, manufacturing, and testing of regulated products. They also establish policies intended to achieve consistency in the Agency's regulatory approach and establish inspection and enforcement procedures. Because guidances are not regulations or laws, they are not enforceable, either through administrative actions or through the courts. An alternative approach may be used if such an approach satisfies the requirements of the applicable statute, regulations, or both. For information on a specific guidance document, please contact the originating office. They includes… · Generics
  • 23. · Generics (Draft - Distributed for comment purposes only) . · Procedural Draft: Applications Covered by Section 505(b)(2) (Issued 10/1999, Posted 12/7/1999). This provision permits FDA to rely, for approval of an NDA, on data not developed by the applicant. · Biopharmaceutics. · Bioavailability and Bioequivalence Studies for Orally Administered Drug Products - General Considerations (Issued 10/2000, Posted 10/27/2000). This guidance should be useful for applicants planning to conduct bioavailability (BA) and bioequivalence (BE) studies during the IND period for an NDA, BE studies intended for submission in an ANDA, and BE studies conducted in the post-approval period for certain changes in both NDAs and ANDAs. · Drug Master Files. A Drug Master File (DMF) is a submission to the FDA that may be used to provide confidential detailed information about facilities, processes, or articles used in the manufacturing, processing, packaging, and storing of one or more human drugs. · Required Specifications for FDA's IND, NDA, and ANDA Drug Master File Binders · Guidance for Industry: Changes to an Approved NDA or ANDA · Refusal to Receive. (Issued 7/12/1993, Posted 11/26/99) Clarifies CDER's decisions to refuse to receive an incomplete application. · Inactive Ingredient Database. This database contains all inactive ingredients present in approved drug products or conditionally approved drug products currently marketed for human use. Laws, Regulations, Policies and Procedures: The mission of FDA is to enforce laws enacted by the U.S. Congress and regulations established by the Agency to protect the consumer's health, safety, and pocketbook. The Federal Food, Drug, and Cosmetic Act is the basic food and drug law of the U.S. With numerous amendments it is the most extensive law of its kind in the world. The law is intended to assure consumers that foods are pure and wholesome, safe to eat, and produced under sanitary conditions; that drugs and devices are safe and effective for their intended uses; that cosmetics are safe and made from appropriate ingredients; and that all labeling and packaging is truthful, informative, and not deceptive. Code of Federal Regulations (CFR): The final regulations published in the Federal Register16 (daily published record of proposed rules, final rules, meeting notices, etc.) are collected in the CFR. The CFR is divided into 50 titles which represent broad areas subject to Federal regulations. The FDA's portion of the CFR interprets the Federal Food, Drug and Cosmetic Act and related statutes. Section 21 of the CFR contains most of the regulations pertaining to food and drugs. The regulations document most actions of all drug sponsors that are required under Federal law. The following regulations apply to the ANDA process: · (21CFR Part 314) Applications for FDA Approval to Market a New Drug or and Antibiotic Drug · (21CFR Part 320) Bioavailability and Bioequivalence Requirements. For more information on retention samples, please see Bioequivalence Study Retention Samples. · Bioavailability and Bioequivalence Requirements; Abbreviated Applications; Final Rule. (Issued and posted 12/19/2002) · (21CFR Part 310) New Drugs MaPPs :- CDER's Manual of Policies and Procedures (MaPPs) provide official instructions for internal practices and procedures followed by CDER staff to help standardize the drug review process and other activities, both internal and external. MaPPs define external activities as well. ANDA review process:
  • 24. Time frames for reviewing ANDA by FDA: Within 180 days of receipt of an application for a new drug under section 505(b) of the act or an abbreviated application for a new drug under section 505(j) of the act, FDA will review it and send the applicant either an approval letter under 314.105 or a complete response letter under 314.110. This 180-day period is called the "initial review cycle." At any time before approval, an applicant may withdraw an application under 314.65 or an abbreviated application under 314.99 and later submit it again for consideration. The initial review cycle may be adjusted by mutual agreement between FDA and an applicant or as provided in 314.60 and 314.96, as the result of a major amendment. Filing of ANDA: Within 60 days after FDA receives an application, the agency will determine whether the application may be filed. The filing of an application means that FDA has made a threshold determination that the application is sufficiently complete to permit a substantive review. If FDA finds that none of the reasons for refusing to file the application apply, the agency will file the application and notify the applicant in writing. The date of filing will be the date 60 days after the date FDA received the application. The date of
  • 25. filing begins the 180-day period described in section 505(c) of the act. This 180-day period is called the "filing clock." Approval of ANDA: FDA will approve an application and issue the applicant an approval letter on the basis of draft labeling if the only deficiencies in the application concern editorial or similar minor deficiencies in the draft labeling. Such approval will be conditioned upon the applicant incorporating the specified labeling changes exactly as directed, and upon the applicant submitting to FDA a copy of the final printed labeling prior to marketing. Amendments to an unapproved ANDA: · An applicant may amend an abbreviated new drug application that is submitted, but not yet approved, to revise existing information or provide additional information. Amendments containing bioequivalence studies must contain reports of all bioequivalence studies conducted by the applicant on the same drug product formulation, unless the information has previously been submitted to FDA in the abbreviated new drug application. A complete study report must be submitted for any bioequivalence study upon which the applicant relies for approval. For all other bioequivalence studies conducted on the same drug product formulation, the applicant must submit either a complete or summary report. If a summary report of a bioequivalence study is submitted and FDA determines that there may be bioequivalence issues or concerns with the product, FDA may require that the applicant submit a complete report of the bioequivalence study to FDA. · Submission of an amendment containing significant data or information before the end of the initial review cycle constitutes an agreement between FDA and the applicant to extend the initial review cycle only for the time necessary to review the significant data or information and for no more than 180 days. · The applicant shall submit a field copy of each amendment. The applicant, other than a foreign applicant, shall include in its submission of each such amendment to FDA a statement certifying that a field copy of the amendment has been sent to the applicant's home FDA district office. References: · Richard A. Guarino- New Drug Approval process-1)The New Drug Application, Content, Format 2) Abbreviated $ Supplementary New Drug Application- Fourth edition-Marcel Dekker,inc- page no 113-183. · Douglas J. Pisano, David S. Manlus –FDA Regulatory Affairs, A guide for Prescription Drugs, Medical Devices and Biologics-New drug Application –Second edition-Marcel Dekker,inc- page no 69-108. · Loyd V. Allen Jr, Nicholas G. Popovich, Howard C. Ansel’s Pharmaceutical Dosage Forms and delivers systems- New Drug Development and Approval Process-8th edition- B.I. publication- Page no 25-65. · http://www.fda.gov/cder/guidance/index.htm . · http://www.fda.gov/drugs/developmentapprovalprocess/howdrugsaredevelopedandapproved/approv alapplications/abbreviatednewdrugapplicationandagenerics/default.htm · http://www1.pointcross.com/source/ddg/charts/andachart.htm · http://www.accessdata.fda.gov/script s /cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=314.94 Common technical document: The common technical document was agreed in November 2000 in sandicgo USA. It provides for a harmonized structure and format for new product applications. This is one in a series of guidance that provide recommendations for applicants preparing the CTD for the registration of pharmaceuticals used for human use, that is , submission to the USFDA . It will significantly reduce time resources. This applicant have used many different approaches to organize the information, but differences in organization of each application made regulatory agency review, more difficult and may lead to omission of critical data or analysis. Such omission can result in a decision by regulatory agency which is not correct for a given drug.
  • 26. So, as to overcome this “US federal drug administration ensured that the worldwide pharmaceutical industry should follow tightly regulated strict standard document maintained in a common format from initial project definition to product approval. · In addition exchange of regulatory information among regulatory authorities will be simplified. · The FDA characterized the CTD as “ an information package of clinical, non clinical and manufacturing, technical data in the same format and with the same content , that would be submitted for registering new drugs in all three ICH the US, European union of Japan. · European Union has its own requirements for the organization of technical reports in submission and for the preparation of the summaries and tables. · In Japan the applicants must prepare the GAIYO which organizes and presents a summary of the technical information. · In Europe experts reports and tabulated summaries are required and return summaries are recommended. · In US FDA has guidance regarding the format and content of new drug application submission. Guidance for industry on preparing the CTD: It has been divided into 4 guidance document on harmonized format. · The organization of the CTD · The quality section · The efficacy section · The safety section Human use and has been subject to consultation by the regulatory parties in accordance with the ICH process. · General considerations for submitting marketing applications according to ICH/CTD format. · CTD guidance’s are intended to be used together with other ICH and agency guidance’s · The guidance suggest that controlled procedure need to be implemented, to specific address – maintenance, storage, access restriction, technical approach, and responsibilities of personal relevant tasks across the retention period of an electronic record. Preparing of organizing the CTD: · The guidance address the general organization of the information to be presented in CTD application for new pharmaceuticals(including biotechnology derived products) · Guidance’s also are available that discuss the quality, efficacy and safety section of the CTD in respective modules. · Applicants should not modify the overall origination of the CTD However in the non clinical and clinical summarizes section of the CTD, applicants can modify individual formats, to provide the best possible presentation of the technical information and to facilitate the understanding evaluation of the results Throughout the CTD, the display of information should be unambiguous and transparent to facilitate the review of the basic data and to help a reviewer become quickly oriented to application contents · Text and tables should be prepared using margins’ that allow the document to be printed on both A4 paper(EU and Japan ) 8.5×1111 paper(US) · The left hand margin should be sufficiently large that information is not obscured through binding. · Font sizes for text and tables should be of a style and size that are large enough to be easily legible, even after photocopying. · 12. Point Font is required for narrative text
  • 27. · Acronyms and abbreviations should be defined the first time they are used in each module. The Common Technical Document (CTD) is a set of specification for application dossier for she registration of Medicines and designed to be used across Europe, Japan and the United States. It was developed by the European Medicines Agency (EMEA, Europe), the Food and Drug Administration (FDA, U.S.) and the Ministry of Health, Labor and Welfare (Japan). The CTD is maintained by the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH).[1] The Common Technical Document is divided into five modules: Administrative and prescribing information Overview and summary of modules 3 to 5 Quality (pharmaceutical documentation) Safety (toxicology studies) Efficacy (clinical studies) Detailed subheadings for each Module are specified for all jurisdictions. The contents of Module 1 and certain subheadings of other Modules will differ, based on national requirements. After the United States, European Union and Japan, the CTD has been adopted by several other countries including Canada and Switzerland. The Paper CTD is destined to be replaced by its electronic counterpart, the eCTD. Organisation of the Common Technical Document The Common Technical Document is organized into five modules. Module 1 is region specific. Modules 2, 3, 4, and 5 are intended to be common for all regions. Conformance with this guideline should ensure that these four modules are provided in a format acceptable to the regulatory authorities. Module 1. Administrative Information and Prescribing Information This module should contain documents specific to each region; for example, application forms or the proposed label for use in the region. The content and format of this module can be specified by the relevant regulatory authorities. Module 2. Common Technical Document Summaries Module 2 should begin with a general introduction to the pharmaceutical, including its pharmacologic class, mode of action, and proposed clinical use. In general, the Introduction should not exceed one page. Module 2 should contain 7 sections in the following order: CTD Table of Contents CTD Introduction
  • 28. Quality Overall Summary Nonclinical Overview Clinical Overview Nonclinical Written and Tabulated Summaries Clinical Summary The organisation of these summaries is described in Guidelines for M4Q, M4S, and M4E. M4-Q: the CTD for the registration of pharmaceuticals for human use –quality ·The quality section of the CTD provides a harmonized structure and format for presenting CMC (chemistry, manufacturing, controls) information in a registration dossier. ·The table of contents include sections on drug substance and drug product. ·Due to the fact that many CMC topics have not yet been the subject of ICH guidelines (e.g.: drug substance synthesis, drug product manufacture), contain the content of CTD-Q is not totally harmonised. ·A new section on pharmaceutical development and also a new CMC summary document, the quality overall summary has been developed. M4-S: the CTD for registration of pharmaceuticals for human use- safety Non –clinical summaries and organisation of module-4 · The M4S guideline will outline the structure of format of nonclinical summaries in module2 of the CTD and will provide the organisation of module 4 i.e. the non-clinical study reports. · The non-clinical overview should present an should present an integrated and critical assessment of the pharmacological, pharmacokinetic, and toxicological evolution of the pharmaceutical and generally not to exceed 30 pages. · The non-clinical written summaries (100-150 pages) are to provide more extensive summaries and discussion of non clinical information on pharmacological, pharmacokinetics and toxicology. M4-E: the CTD for the registration of pharmaceuticals for human use-efficacy Module-2:clinical overview and clinical summary Module -5:clinical study reports. M4E describes the structure and format of the clinical data in an application, including summaries and detailed study reports. There are two high level clinical summaries in module 2 of the CTD: Clinical overview: a short document that provides a critical assessment of the clinical data Clinical summary: a longer document that focuses on data summarisation and integration. Module 3. Quality Information on Quality should be presented in the structured format described in Guideline M4Q. Module 4. Nonclinical Study Reports The nonclinical study reports should be presented in the order described in Guideline M4S. Module 5. Clinical Study Reports The human study reports and related information should be presented in the order described in Guideline M4E. Organisation of the Common Technical Document for the Registration of Pharmaceuticals for Human Use Module 1: Administrative Information and Prescribing Information Table of Contents of the Submission Including Module 1 Documents Specific to Each Region (for example, application forms, prescribing information) Module 2: Common Technical Document Summaries Common Technical Document Table of Contents (Modules 2-5) CTD Introduction
  • 29. Quality Overall Summary Nonclinical Overview Clinical Overview Nonclinical Written and Tabulated Summaries Pharmacology Pharmacokinetics Toxicology Clinical Summary Biopharmaceutical Studies and Associated Analytical Methods Clinical Pharmacology Studies Clinical Efficacy Clinical Safety Literature References Synopses of Individual Studies Module 3: Quality Table of Contents of Module 3 Body of Data Literature References Module 4: Nonclinical Study Reports Table of Contents of Module 4 Study Reports Literature References Module 5: Clinical Study Reports Table of Contents of Module 5 Tabular Listing of All Clinical Studies Clinical Study Reports Literature References Reference: ·Guidance for Industry, ICH M4: Organization of the CTD" U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER) Center for Biologics Evaluation and Research (CBER) August 2001 SUPAC ( SCALE - UP AND POST- APPROVAL CHANGES ) Product development aims at formulating active drug ingredient in a palatable form . Technology transfer of a pharmaceutical product from research to the production floor (shop floor) with simultaneous increase in production outputs is commonly known as scaleup.In simple terms the process of increasing batch size is termed as scaleup.Scale down refers to decrease in batch size in response to reduced market requirements. SUPAC refers to the FDA recommended testing and filling actions to be taken by a pharmaceutical firm when it changes the manufacturing process of a drug product that has been approved via a New Drug Application(NDA),an Abbreviated New Drug Application (ANDA),or an Abbreviated Antibiotic Drug Application(AADA). It is designed to lower the regulatory burden associated with alterations in batch size ,components and composition ,manufacturing site or manufacturing of a marketed product. A drug may experience many changes during its life cycle .These changes may have an adverse effect on the overall safety and effectiveness of the drug or drug product .After a number of changes over a long time period ,the product coming to market maybe completely different from the one that was approved.Hence data submitted to regulatory authorities in support of a change must have a comparison record of the drug or drug product to the one that was approved initially.documentation generated in support of any change to
  • 30. the approved drug is submitted to regulatory authority for review,and based on the benefit to risk ratio ,the drug or drug product is approved.depending upon the intensity of change ,supporting documents are provided to the regulatory agency. The SUPAC Guidance offer an advantage over the regulations that the documents are specific for particular dosage forms.This approach was taken since some product types are more complicated than others,and require more complicated controls.Two guidance have been finalized.They are : 1. Immediate release solid oral dosage forms –Scale up and post approval changes :Chemistry,Manufacturing and controls,in vitro dissolution testing ,and invivo bioequivalence documentation. 2. Non sterile semisolid dosage forms –Scaleup and post approval changes : :Chemistry, Manufacturing and controls, in vitro dissolution testing ,and invivo bioequivalence documentation. In addition ,SUPAC documents covering other dosage forms (eg, extended-release products,transdermals,parenteral solutions)as well as a related documents for bulk active substances ,are at various stages of development. Modifications are divided into one of three categories, each with specific testing and documentation requirements. Level 1 changes: Are those that are unlikely to have any detectable impact on formulation quality or performance ,such as change of less than 5% in the concentration of an excipient. likely to have a significant impact on quality or performance. SUPAC Stability Testing: In addition to in vitro release testing ,longterm stability studies are required at all levels of change ,while accelerated stability studies are required for levels 2 and 3. SUPAC Testing : The purpose of SUPAC Testing is to verify that there is no change in product quality or performance after a modification .A key component of a level 2 or 3 change is in vitro release testing for pre and post change formulations. For SUPAC Submissions,the following points should also be considered : 1.In most cases, except those involving scale-up,stability data from pilot scale batches will be acceptable to support the proposed change. 2.Where stability data show a trend towards potency loss or degradant increase under accelerated conditions ,it is recommended that historical accelerated stability data from a representative prechange batch be submitted for comparision. An invitro release rate can reflect the combined effect of several physical and chemical parameters ,including solubility and particle size of the active ingredient and rheological properties of the dosageform.In most cases,invitro release rate is a useful test assess between prechange and post change products. 1.In vitro release testing is a useful test to assess product “sameness”under certain scaleup and post approval changes for semisolid products. 2.The development and validation of an invitro release test are not required for approval of an NDA,ANDA,AADA nor is the invitro release test required as a routine batch to batch quality control test. 3.In vitro release testing ,alone ,is not a surrogate test for in vivo bioavailability or bioequivalence. 4.The in vitro release rate shouldnot be used for comparing different formulations across manufacturers. The SUPAC guidance documents defines 1)levels of change 2)recommended chemistry,manufacturing,and controls tests for each level of change. 3)In vitro dissolution tests and in vivo bioequivalence tests for each level of change. 4)documentation that should support the change for new drug applications(NDA)and abbreviated new drug applications(ANDAs).
  • 31. This guidance provides recommendations to sponsors of new drug applications(NDA),abbreviated antibiotic applications(AADA’s),who intend during the post approval period,to change :1)The components or composition:2)the site of manufacture3)The scale up/scale down of manufacture 4)the manufacturing(process and equipment)of an immediate release oral formulations. The purpose of this guidance is to provided recommendations to pharmaceutical Manufacturers using the Center for Drug Evaluation and Research’s guidance for Industry: Immediate Release Solid Oral Dosage Forms-Scale up and post approval changes: Chemistry, manufacturing and controls ,In vitro Dissolution testing ,and in vivo Bioequivalence Documentation(SUPAC-IR) ,which is published in November 1995, and guidance for industry. Dissolution testing: Case A: Dissolution of Q=85%in 15min in 900milliliters of 0.1N hydrochloride using the United States Pharmacopoeia. Apparatus 1 at 100 revolutions per minute or Apparatus 2 At 50 rpm. Case B: Multi point dissolution profile in the application/compendial medium at 15,30,45,60,120min until an asymptote is reached for the proposed and currently Accepted formulation. Case C: Multi point dissolution profiles performed in water ,0.1N HCL,and USP buffer media at pH 4.5,6.5,7.5 for the proposed and currently accepted formulations. Adequate sampling should be performed at 15,30,45,60,120 min until either 90% of drug from the drug product is dissolved or an asymptote is reached .A surfactant maybe used with appropriate justification. Components and composition: It focuses on changes in excipients in the drug product Changes in the amount of drug substance are not addressed by this guidance Changes in components or composition that have the effect of adding a new excipient or deleting an excipient. A.Level 1 changes. 1.Definition of level Level 1 changes are those that are unlikely to have any detectable impacts on formulation quality and performance. Examples: a.Deletion or partial deletion of an ingredient intended to affect the color or flavor of the drug product ;or change in the ingredient of the printing ink to another approved ingredient . b.changes in excipients ,expressed as percentage (w/w)of total formulation ,less than or equal to the following percent ranges: Excipient percent excipient(w/w)out of total target dosage form weight Filler ± 10 Disintigrant-starch ±6 : other ± 2 Binder ± 1 Lubricant –calcium or magnesium stearate ± 0.5 Other ± 2 Glidant - talc ±2 other±0.2 filmcoat- ±2 These percentages are based on the assumption that the drug substance in the drug product is formulated to 100%of label /potency .The total additive effect of all excipient changes should not change by more than 10%. 2. Test documentation a. Chemistry documentation: Application /compendial release requirements and batch records. Stability testing:one batch on long term stability data reported in annual report. b.Dissolution documentation c.Invivo Bioequivalence documentation
  • 32. 3.Filing documentation-prior approval supplement(all information including accelerated stability data);annual report(long term stability data). Level 2 changes: are those that could have a significant impact on formulation quality and performance. Tests and filing documentation for a level 2 change vary depending on three factors :therapeutic range,solubility,permeability. Level 3 changes:are those that are likely to have a significant impact on formulation quality and performance.Tests and filling documentation vary depending on the 3 factors;therapeutic range ,solubility,permeability.. SITE CHANGES: Site changes consist of changes in location of the site of manufacture for both company-owned and contract manufacturing facilities and donot include any scaleup changes ,changes in manufacturing(including process and/or equipment),or changes in components or composition .New manufacturing locations should have a satisfactory current Good manufacturing practice (CGMP)inspection.. A.Level 1 changes 1.Definition of level Level 1 changes consists of site changes with in a single facility where the same equipment ,standard operating procedures ,environmental conditions sites are used,and where no changes are made.and controls and personnel common to both manufacturing to the manufacturing batch records except for administrative information and the location of the facility.common is defined as employees already working on the campus who have suitable experience with the manufacturing process. 2.Test documentation a)Chemistry documentation None beyond application/compendial release requirements. b)Dissolution documentation None beyond application/compendial release requirements. c)Invivo bioequivalence documentation None 3.Filing documentation Annual report B.Level 2 changes 1.Definition of level Level 2 changes consists of site changes with in a contiguous campus,or between facilities in adjacent city blocks,where the same equipment,SOP’s,environmental conditions(eg temparature and humidity)and controls,and personnel common to both manufacturing sites are used ,and where no changes are made to the manufacturing sites are used ,and where no changes are made to the manufacturing batch records,except for administrative information and the location of the facility. 2.Test documentation a.Chemistry Documentation Location of new site and updated batch records.None beyond application /compendial release requirements. One batch on long term stability data reported in annual report. b.Dissolution Documentation None beyond application /compendial release requirements. c.Invivo Bioequivalence documentation None 3.Filing documentation Changes being effected supplement :annual report(long term stability test data) C.Level 3 changes 1.Definition of level
  • 33. Level 3 changes consists of a change in manufacturing site to a different campus.A different campus is defined as one that is not on the same original contiguous site or where the facilities are not in adjacent city blocks.To qualifyas a level 3 change,The same equipment ,SOP’s ,environmental conditions ,and controls shouldbe used in the manufacturing process at the new site ,and no changes maybe made to the manufacturing batch records except for administrative information ,location,language translation,where needed. 2.Test documentation a)Chemistry documentation Location of new site and updated batch records. Application/compendial release requirements . Stability: Significant body of the data available: One batch with three months acclerated stability data reported in supplement;one batch on longterm stability data reported in annual report. Significant body of the data not available: Up to three batches with three months acclerated stability data reported in supplement;up to three batches on long term stability data reported in annual report. b)Dissolution documentation CaseB:multi point dissolution profile should be performed in the application/compendial medium at 15,30,45,60,120 minutes or until an asymptote is reached. The dissolution profile of the drug product at the current and proposed site should be similar. c) Invivo Bioequivalence Documentation: None 3. Filing Documentation Changes being effected supplement;annual report(long term stability data). CHANGES IN BATCH SIZE (SCALE- UP/SCALE DOWN) : Post approval changes in the size of a batch from the pivotal/pilot scale batch material to larger or smaller production batches call for submission of additional information in the application. Scale down below 100,000 dosage units is not covered by this guidance .All scale-up changes should be properly validated and, where needed, inspected by appropriate agency personnel. A)Level 1 changes 1. Definition of level Change in batch size up to and including a factor of 10 times the size of the pilotbio batch where; i) The equipment used to produce the test batches is of the same design and operating principles; ii) The batches is manufactured in full compliance with CGMP’s. iii) The same standard operating procedure(SOP’s)and controls, as well as the same formulation and manufacturing procedures, are used on the test batches and on the full scale production batches 2. Test documentation a)Chemistry documentation Application/Compendial release requirements .Notification of change and submission of updated batch records in annual report. One batch on long term stability reported in annual report . b)Dissolution documentation None beyond application /compendial release requirements. c)In vivo Bio equivalence None 3.Filing Documentation Annual report (long term stability data). B)Level 2 changes
  • 34. 1)Definition of level Changes in batch size beyond a factor of ten times the size of the pilot/biobatch,where; i)The equipment used to produce the test batches is of the same design and operating principles; ii)The batches is manufactured in full compliance with CGMP’s. iii)The same standard operating procedure(SOP’s)and controls, as well as the same formulation and manufacturing procedures,are used on the test batches and on the full scale production batches. 2.Test documentation a)Chemistry documentation Application/compendial release requirements .Notification of change and submission of updated batch records. Stability testing: One batch with three months accelerated stability data and one batch on longterm stability. b)Dissolution documentation Case B testing c)In vivo Bioequivalence documentation None. 3.Filing Documentation Changes being effected supplement :annual report(long term stability data) MANUFACTURING CHANGES: Manufacturing changes may affect both equipment used in the manufacturing process and the process itself. A.Equipment 1.Level 1 changes a)Definition of change This category consists of : 1)Change from non-automated or non mechanical equipment to move ingredients:and 2)Change to alternative equipment of the same design and operating principles of the same or of a different capacity. b)Test documentation i)Chemistry documentation Application/compendial release requirements .Notification of change and submission of updated batch records. Stability testng:One batch on longterm stability. b)Dissolution documentation None beyond application/compendial release requirements. c)In vivo Bioequivalence documentation None. c)Filing documentation Annual report(Long term stability data 2.Level 2 Changes a)Definition of level Change in equipment to a different design and different operating principles. b)Test documentation i)Chemistry documentation Application/compendial release requirements .Notification of change and submission of updated batch records. Stability testing Significant body of data available: One batch with three months accelerated stability data reported in supplement ;One batch on long term stability data reported in annual report.
  • 35. Significant body of data not available Up to three batches with three months accelerated stability data reported in supplement;upto three batches on long term stability data reported in annual report. ii)Dissolution documentation Case C dissolution profile. iii)Invivo Bioequivalence documentation None c.Filing documentation Prior approval supplement with justification for change ;annual report(long term stability data). B)Process a)Definition of level This category includes process changes including changes such as mixing times and operating speeds within applicaton/validation ranges. b)Test documentation i)Chemistry documentation None beyond application/compendial release requirements. ii)Dissolution documentation None beyond application/compendial release requirements iii)In vivo Bioequivalence documentation None c).Filing documentation Annual report 2.Level 2 changes a.Definition of level This category includes process changes including changes such as mixing times and operating speeds outside of application / validation ranges. b)Test documentation i)Chemistry documentation Application/compendial release requirements .Notification of change and submission of updated batch records. Stability testing:one batch on longterm stability. ii)Dissolution documentation Case B dissolution profile. iii)Invivo Bioequivalence documentation None c).Filing documentation Changes being effected supplement ;annual report(long term stability data). 3.Level 3 changes a).Definition of level This category includes changes in the type of process used in the manufacture of the product ,such as a change from wet granulation to direct compression of dry powder. b)Test documentation i)Chemistry documentation Application/compendial release requirements .Notification of change and submission of updated batch records. Stability testing: Siignificant body of data available: One batch with three months accelerated stability data reported in supplement ;one batch on long term stability data reported in annual report. Significant body of data not available:
  • 36. Up to three batches with three months accelerated stability data reported in supplement ;up to three batches on long term stability data reported in annual report. ii)Dissolution documentation case B dissolution. iii)Invivo Bioequivalence documentation Invivo bioequivalence study .The bioequivalence study maybe waived if a suitable in vivo/invitro correlation has been verified. c).Filing documentation Prior approval supplement with justification ;annual report (long term stability data). Handling and Maintenance of Electronic documentation Introduction of Electronic documentation: An electronic document is any electronic media content (other than computer programs or system files) that is intended to be used in either an electronic form or as printed output. Originally, any computer data were considered as something internal the final data output was always on paper. However, the development of computer networks has made it so that in most cases it is much more convenient to distribute electronic documents than printed ones. And the improvements in electronic display technologies mean that in most cases it is possible to view documents on screen instead of printing them (thus saving paper and the space required to store the printed copies). However, using electronic documents for final presentation instead of paper has created the problem of multiple incompatible file formats. Even plain text computer files are not free from this problem e.g. under MS-DOS, most programs could not work correctly with UNIX-style text files (see newline), and for non- English speakers, the different code pages always have been a source of trouble. Even more problems are connected with complex file formats of various word processors, spread sheets and graphics software. To alleviate the problem, many software companies distribute free file viewers for their proprietary file formats (one example is Adobe's Acrobat Reader). The other solution is the development of standardized non-proprietary file formats (such as HTML and Open Document), and electronic documents for specialized uses have specialized formats–the specialized electronic articles in physics use TeX or PostScript. GUIDELINES FOR RECORDS OF LONG-TERM OR ENDURING LEGAL VALUE STORED ON ELECTRONIC MEDIA Planning Process: Access: Before you convert your information system, decide how you will provide ready access to the records you store off-line. Media and systems: Select appropriate media and systems for maintaining your record information. Recommended: Magnetic tapes or cartridges are preferred for the long-term storage of electronic records. CD-ROMs created with the ANSI 9660 standard can be used as well. The storage capacity of a magnetic tape, however, far exceeds that of a CD-ROM. Not Recommended: Floppy disks, removable disks and optical disks are not recommended for the long-term storage of electronic records. Maintenance: Access: Maintain your records in a useable format and keep up to date all materials needed to access them, including indexes and other documentation, until they are scheduled for disposal or for transfer to the State Archives. Backups: Maintain in an off-site location backup and archived copies of records and all materials required to access them. Labeling: Develop procedures for labeling tapes and cartridges. Each external label should carry information unique to the tape or cartridge it identifies. At minimum, it should display the name of the organizational unit responsible for the data, the system title, the file title, the destruction date or permanent status of the record, and its security classification, if applicable.