Handbook of Pharmaceutical Generic Development Vol 01 Part 1 (Tab)
Handbook of Pharmaceutical Generic Development Vol 01 Part 1 (Tab)
HANDBOOK OF PHARMACEUTICAL
GENERIC DEVELOPMENT
T ablets
GENERIC DEVELOPMENT
Handbook of Pharmaceutical
Generic Development Series
Handbook of
Pharmaceutical
Generic
Development
Part One
ORAL
Immediate Release
TABLETS
Drug D e v e l o p m e n t
The Complete H a n d b o o k S e r i e s o f
Pharmaceutical Drug Development
ISBN 0793 8632 - Electronic Version
Handbook Development 24 Volume Series
ISSN Series Number 0793 761X - Electronic Version
Available as Print, Online, CD ROM or electronic mail attachment. Additional Drug Specific Volumes in Preparation.
An on-going electronic and print series. Available either as Hard Bound, Soft Bound or Soft Spiral Cover (for Updating).
For Drug Specific Handbooks refer to the 120+ Drug Development Series titled
READY-TO-GO™ DRUG DEVELOPMENT SERIES
http://www.locumusa.com/2go
p p http://www.iagim.org p p
Acknowledgments
I.A.G.I.M. (R&D) Foundation.
I.A.G.I.M. Members (1994 - 2000).
Contributions - Generic & Research Firms
Associate Universities, Technicons and Consultants.
Handbook Series Coordinating Committee.
International Journal of Drug Development.
Journal of Pharmaceutical Development.
International Journal of Generic Drugs.
I.A.G.I.M. Drug Development Archives
Locum International Archives.
FDA/OGD/CDER Maryland
Guides and Guidelines
Library of Congress.
AIC Conferences.
Editorial Board.
Pharm. Eur.
USP/NF.
USPC.
BP.
°
To Doribelle
for her years of support and help
to Sean for his expert knowledge on computerization
to David and Ari for running the project's computers
and lastly to Pat for his inestimable
contribution.
24 Volume Series
Handbook of Generic development
Third International Edition.
L O C U M P U B L I S H I N G H O U S E
Í °Î
Ï Ð
Í °Î
INTRODUCTION
Handbook of Generic Development - Oral Tablet Dosage Form
This handbook is the third international edition of the ongoing 24 volume series
under the cumulative title of Handbook of Generic Drug Development. It is a hands-
on, technical presentation that portrays the current drug requirement steps
necessary at the time of going to print, of the Abbreviated New Drug Application for
oral tablet dosage form, namely tablets and caplets. It is written in conjunction with
Part Two of the Handbook which models as a representative ANDA and as an
example of the drug development process required for solid oral dosage forms The
Handbook is available in electronic format (CD ROM) and e-format (on-line). The
Handbook is up-dated to current regulatory requirements once or occasionally under
exceptional circumstances twice annually. Complete updates are available without
charge to Association Members of the Drug Development Association - IAGIM.
This handbook provides a proven pathway to solid oral dosage form development.
Modern commercial formulations highlight the common tablet/caplet development
routes namely the classical wet granulation, spray granulation, dry granulation and
finally slugging and direct compression. Low active dosage (<10mg) and high
potency (>50%) examples are specially chosen to demonstrate the formulation steps
and process stages as a prerequisite to developing stable, elegant and rugged
formulas.
This Handbook edition includes additional data on analytical method validation has
been redesigned to meet the Guidance for Industry - Organization of an Abbreviated
New Drug Application and an Abbreviated Antibiotic Application as well as all FDA
guideline and requirements of the Center of Drug Evaluation and Research (CDER)
to date of publishing. Editor-in-Chief.
ÓšÎ
© COPYRIGHT 1995-00
pÏ›Ðp
Contents
PHARMACEUTICAL DEVELOPMENT
Chapter 1
Regulatory 1.1
- Pre-formulation checklist 1.3
Documentation 1.4
- SOP Control checklist 1.5
Development Notebooks 1.6
- Development Notebooks checklist 1.7
- SOP Control and Development Notebooks SOPs 1.8
Chapter 2
Developing the Formula -an Overview 2.1
- Formulation checklist 2.2
- Development formulations 2.3
Drug Development Checklist 2.4
Development Formula SOPs 2.5
Developing the Formula 2.6
Product Development Flow Chart 2.11
Product Development Guide 2.13
Direct Compression Tablet Development 2.21
Direct Compression Flow Charts 2.26
Direct Compression Master Formula 2.29
Wet Granulation Development 2.31
Wet & Direct Compression Flow Charts 2.26
Wet Granulation Manufacturing Flow Charts 2.41
Wet Granulation Master Formula (aqueous granulation) 2.42
Wet Granulation Master Formula (alcoholic granulation) 2.44
Contents
Chapter 2
Purified Water - an essential ingredient 2.45
Do and Don'ts in Development 2.48
Purified Water - Checklist 2.49
Chapter 3
Active Ingredients 3.1
-Do’s and Don’ts 3.2
-Active checklist 3.3
-Approved Suppliers Checklist 3.5
-Standard Operating Procedures, Actives 3.6
Chapter 4
Semi active ingredients 4.1
-Validating the Semi-active ingredients, Checklist 4.2
Qualifying the Antioxidant 4.4
Antioxidant Tabulations 4.5
Chapter 5
Non active materials (excipients) 5.1
-Checklist non active ingredient 5.2
-Standard Operating Procedures, Non actives 5.3
Chapter 6
Container closure systems 6.1
-Container-liner-closure systems, Checklist 6.2
-Container-liner-closure systems, SOPs 6.3
-Packaging Components 6.5
-Packaging Components Documentation Requirements SOP 6.6
-Packaging Characteristics 6.11
-Packaging Component Descriptions 6.12
-Packaging Component 6.16
Chapter 7
Manufacturing Instructions 7.1
- Manufacturing Instructions; Checklist 7.4
- The manufacturing Instructions and Controls 7.5
- Manufacturing Flow Charts 7.16
- Large scale manufacturing Instructions 7.20
- Large scale Master Formula 7.24
- Large scale Manufacturing Instructions 7.27
Contents
Chapter 8
In-process Quality Controls 8.1
-Manufacturing in-process controls; Checklist 8.3
-In-process Specifications 8.5
-Process yields 8.8
-In-process Control Specifications 8.9
-In-process Film Coating Specifications 8.10
Chapter 9
Finished Product Specifications 9.1
- Finished Product Specifications Tablet & Caplets 9.2
- Finished Product Specifications; Checklists 9.4
- Finished Product Specifications; Required SOPs 9.5
Chapter 10
Process Optimization and Procedures 10.1
Qualification of Antioxidant and Lubricant 10.2
Qualification of Loss on Drying Limits 10.3
Chapter 11
Scale-up Procedures 11.1
- Scale-up procedures; checklist 11.3
- Scale-up procedures; Aqueous Film Coating 11.6
Chapter 12
Cleaning Limits 12.1
Cleaning Limits Procedures; Checklist 12.6
Cleaning Validation Requirements; SOPs 12.8
Chapter 13
Analytical Validation Requirements 13.1
-Analytical Testing Out of Specification 13.21
-Analytical Testing Do's and Don'ts - Retesting Rules 13.23
-Out-of-Specifications Checklists 13.24
-Ruggedness and Robustness 13.37
-Impurities in Drug Substances 13.41
-Impurities Do's and Don'ts 13.50
-Impurities Glossary of terms 13.51
-Impurities Decision Trees 13.53
Analytical Post approval Changes - PAC/ALTS 13.55
PAC-ALTS Checklist 13.58
Contents
Chapter 14
Process Qualification Batch 14.1
-Process Qualification Batch; Checklist 14.2
-Process Qualification Batch; SOPs 14.3
-Process Qualification Blend Analysis 14.7
-Process Qualification Blend Analysis - Do's and Don'ts 14.7
-Process Qualification Qualifying Tablet Hardness 14.8
Tablet Hardness Protocol 14.10
Chapter 15
Pivotal batch
-The Pivotal Batch 15.1
-Pivotal batch Checklist 15.2
-Pivotal batch SOPs 15.3
-Sampling and Testing the Pivotal Batch 15.3
-Auditing the Pivotal batch 15.4
-Auditing the Pivotal batch Checklist 15.9
Chapter 16
Bioequivalence vs. RLD 16.1
Test Designs - Overview 16.2
Statistical Bioequivalence 16.13
IBE Equation explained 16.14
IBE - Big Picture (Pros and Cons) 16.15
Comparing IBE and ABE 16.16
Dissolution Testing in IR Dosage Forms 16.18
Typical IVIVC Models 16.21
Choosing IVIVC levels 16.22
Dissolution Testing in IR Solid Dosage Forms 16.23
Similarity Factor in dissolution testing 16.30
Biowaivers 16.32
Overall Dissolution Picture 16.35
Biopharmaceutics Classification System 16.36
Evaluating Differences between Drug, Powder Blend, and Tablets 16.37
Performance Verification in Dissolution testing 16.39
Food-Effects in BA-BE Studies 16.52
Similarity Testing - Chow, Pitt and Others 16.60
Contents
Chapter 17
Technical Transfer Documentation 17.1
-Technical Transfer Documentation; Do's and Don'ts Checklist 17.5
-Technical Transfer Documentation; Pharmaceutical Part 17.7
-Technical Transfer Documentation; Analytical Part 17.10
Chapter 18
Process Validation Batches 18.1
-The Process Validation Batches - Essential know-how 18.2
-Process Validation Requirements; SOPs 18.4
-Process Validation Master Plans 18.5
Chapter 19
Pre--Approval Inspections 19.1
PAI Summary 19.8
Pre--Approval Inspection Audit - Team Set Up 19.9
Pre--Approval Inspection Audit - Team Activities 19.11
Chapter 20
Stability Testing of Drug Substance and Drug Product I 20.1
Stability Testing of Drug Substance and Drug Product II 20.15
Stability Testing of Drug Substance and Drug Product II 20.31
Stability Testing Significant Change 20.28
Stability Storage Conditions 20.29
Photostability in Drug Substances 20.31
Setting up a Functional Stability Unit 20.40
Stability SOPs Development 20.48
Chapter 21
Standard Operational Procedures
Development SOPs 21.1
Index of Pharmaceutical Standard Operating Procedures 21.3
Index of Analytical Standard Operating Procedures 21.10
Index of Microbiological Standard Operating Procedures 21.16
Index of Stability Standard Operating Procedures 21.20
Model Development Report 22.1
Experimental Development 22.2
Pre-formulation Development 22.7
Small scale Development 22.9
Scale-up 22.14
Pivotal Lot Manufacture 22.27
Model Development Report - Summary and Conclusion 22.41
HANDBOOK OF GENERIC DRUG DEVELOPMENT XI 24 VOLUME HANDBOOK SERIES
ORAL TABLETS Table of Contents.
ÓšÎ
International Print Edition
p20±00p
© COPYRIGHT 1995-00
Ï›Ð
H P G D
Handbook of Pharmaceutical Generic Development
™
This Print or CD ROM edition of the Drug Development series has been updated to June 2000 Office of Generic
Drug's requirements. Handbook clients requiring to continue this annual ONGOING service need only to
become members of the Drug Development Association (I.A.G.I.M.) for the period of the update service as
required by the firm, starting from date of purchase. The Drug Development Update Program is renewed in
December each year as a function of the firms requirements. New Drug Formula And Manufacturing Procedures
Are Added Annually All updates are on CD ROM in PDF™. Warning: Copyright © 1985 -2000 Locum
Publishing House Inc.-All Rights Reserved.
Neither this information or nor any part of the data contained therein may be reproduced, copied or transmitted
in any form, modification or merged portion or by any means, electronic or mechanical, including printing
photocopying, microfilming and recording, or by any information storage and retrieval system, without the prior
written permission of the publishers. ™ Trademark - Locum Corporation, ™ Locum International Group.
http://www.locum.co.il
[email protected] - [email protected]
http://www.locumusa.com
[email protected] - [email protected]
http://www.locumeuro.com
(See web site for IAGIM Application Membership Forms)
[email protected]
http://www.iagim.org
[email protected] - [email protected]
http://www.locumusa.com
[email protected] - [email protected]
http://www.locumeuro.com
Introduction
T
he purpose of the Handbook Series is to illustrate generic drug development from pre-
formulation to regulatory submission. The Handbook Series on pharmaceutical dosage
forms deals with the US generic drug development process of the ANDA (Abbreviated
New Drug Application) and EU Dossier It is well suited to innovative development
processes for the Chemistry-Manufacturing-Controls (CMC) Section of an New Drug
Application (NDA).
Each book is devoted to a specific dosage form e.g. tablets, capsules, (immediate & modified
release), liquids, topical semi solids, suspensions, eye and nose preparations, inhalation
aerosols and so forth. It is an ongoing series that is reviewed and updated twice annually, as
new agency regulations, guides, guidelines and industry procedures are adopted or regulated.
The Handbook is a basic hands-on working approach to generic drug development and the
overall developmental process. The handbook ends with the requirements for manufacturing
the first three commercial product lots for distribution and marketing.
Each Handbook is presented in two volumes referred to as Part One and Part Two. These two
parts are supplementary and should be used and referenced together, as they complement
each other. Electronic templates for the full registration process are available for each dosage
form. These approximate +300 Word™ based templates consist of electronically completed
ANDA data where only the variable facts and figures need to be inserted into the prepared
data fields. Templates may be incorporated readily into any popular documentation system
Part One covers the development topics from pre-formulation of generic ANDAs to final FDA
filing with the Office of Generic Drugs. Each chapter details key development steps coupled
with a hands-on development checklists and specification that dovetail with a series of SOPs
on practical generic issues that the FDA review chemists and inspectors routinely address
during an ANDA file review and during pre-approval site inspections (PAIs). Agency site
inspections routinely cover the product development unit or R&D departments, QC and
Analytical Research laboratories, as well as the manufacturing facilities and production
warehouses. During a product-specific pre-approval inspection there is a concentration of
effort by the inspectorate to thoroughly review and evaluate the drug development process
from pre-formulation to pivotal batch.
Topics covered are real life examples from A (actives) to V (validation). Procedures are kept
as simple as possible in order that the checklists and SOPs can be understood by all
departmental personnel concerned. Specifications checklists and SOPs used emphasize
essential procedures or requirements meeting all the published FDA guides and guidelines at
the time of publication Thus the checklists become a first party audit or self-inspection format
for the Standard Operating Procedures.
Part Two is a complete real-life dosage form specific working model of a US (or EU) Generic
Application In the US, it is commonly known as an Abbreviated New Drug Application (ANDA).
In Part Two, the model ANDA (or a model EU dossier) meets the current agency expectations
on content, file assembly and format - the manufacturing data in chapters 11 and 12 are taken
from commercial processes currently in practice as presented in the Ready-to-Go™ 120+ Drug
development know-how series.
3
Preface
“… getting a generic drug to the market place on time….”
G
etting a generic drug to the market place at the right time is no easy task. The
generic drug product must be approved by the FDA close to the latest patent
and exclusivity expiration date of the innovator drug, if a firm wants to be the
first generic drug product on the retail shelves.
Furthermore the three commercial validation batches should be manufactured, filled
and packed via a full-scale standard production run. The now ready-to-launch generic
drug product must meet all its product specifications and the three commercial
validation lots should be on and real time stability evaluation for at least one to three
months. Should all this work have been completed on time and the manufacturing
facility is in full GMP compliance with all manufacturing and control documentation in
place - then the generic product has been developed ‘on time’.
Getting to this point is a long training and planning operation. That it can be done has
been shown by dedicated and well managed generic-innovative and generic
companies. This handbook is designed to show the key highlights of the essential
training and planning along the way.
It is not a manual on how to pre-formulate or formulate a specific dosage form, more
over it is a handbook on how to plan, manage and deliver all the key ingredients of a
successful generic drug product from pre-formulation to the marketed generic drug -
on time and without a delay in the drug product development process.
The length and breath and importance of preparing a successful long life generic
product for the market place requires much attention to detail. Development must stop
if the product fails an essential intermediate, finished product or stability specification
and continue only after the fault has been isolated and corrected - thus the essential
use of checklists and standard operating procedures in this Handbook. The SOPs are
generic in content, they simply highlight important principles and way points and are
suitable for editing and customizing for the firms own in-house needs.
The FDA file contents and review expectations of the drug product must be well
understood and controlled early in the development process in order to avoid problems
with the approval process and later with the quality and customer acceptance of the
marketed product.
This Handbook emphasizes ‘first party certification’ by in-house auditing and self
inspection programs exhibiting a past systematic QC track record that may help
streamline FDA’s enforcement of drug Good Manufacturing Practices (GMPs).
This handbook was designed to produce a rugged generic drug product - to rapidly
facilitate FDA and pre-approval review and reach the market place on time...
Editor
in an HPLC assay spectrum, was found That regulator won't forget you or your
on investigation to arise from a change product line up for review!
in an inactive dye vendor (a new Work with regulators - or they will work
supplier) and not as was anticipated a against you and your product may not
new product impurity or degradant. get to the market place on time.
Directive 8. Treat regulators with respect - as you
"Run a mock PAI against your would like to be treated. Agency official
Application just before submitting." are understanding, experienced
The Drug Application will eventually be professionals whose prime concern is
judged on the acceptability of the product quality and safety.
manufacture, control and testing In any regulatory meeting the only
facilities as documented in the agency
welcome outcome is a win-win scenario.
file and in-house supporting data. Audit
Both parties get what they want.
every facet of the development,
manufacture, control and stability Remember an agency never looses an
procedures of the drug product. Check argument - the product only suffers and
and cross-reference each possible gets delayed due to incomplete data or
submission document against the regulatory requirements.
manufacturing / control and laboratory Directive 11.
files and equipment logs. Build in routine "Talk to the regulators regularly."
self-inspection checks during the Allow regulators to review protocols prior
development process. Formulate this to starting the work. Get their opinion
quality development routine by SOPs and express your concerns openly.
and department audit checklists. Regulators like openness and honesty -
Directive 9. and work well with polite, respectful and
"Make your Application really clear, professional personnel.
concise and user friendly " Directive 12.
Well prepared and assembled print or "Take a hard look a your cGMP's "
electronic files and dossiers are a joy to
The absence of GMP compliance simply
read, review, and evaluate. Use all the
adulterates your drug development
desk top publishing tools to shape your
pipeline. GMP compliance is targeted to
firm's reports as attractive, stimulating,
play a more dynamic role in the drug
and interesting to read and review.
review and PAI process.
A document can entice or repel a reader (Establishment Evaluation System - FDA Drug Center and
simply by its construction - it can also be Office of Regulatory Affairs electronic data sharing)
made a scientific work-of-art. Directive 13.
Directive 10. "Audit everything enthusiastically".
"Treat regulators like your key Leave no audit stone unturned.
personnel treat you" Establish consistent in-house audit self-
Listen to regulators - they too have their inspection programs effective at each
story to tell and may know regulations stage of the drug development pathway.
that you don't. Listen to their concerns At the end of every development report
clearly - it's in your product's interest. submission stage, (refer development
There is no greater achievement in checklist chapter 15), audit the department
satisfying a PAI inspector's requests in and relevant steps concerned.
real-time and in producing the End-of-study auditing is quite ineffective
documentation/data requested - before as early errors or omissions can not be
he leaves the firms' premises. corrected promptly and on-time. 2
Regulatory
‘Sit and review with your regulatory department before you start’
D eveloping successful generic drugs in the least possible time and without
expensive mistakes, requires significant pre-planning with the regulatory affairs
department. Successful generic project managers can visualize all the key
sections of the ANDA submission file, before pre-formulation work has actually been
initiated.
The regulatory department must insure that the innovators’ drug or the reference
listed drug ( R L D ) is suitable for generic manufacture and marketing.
This purpose of this Handbook is to allow the reader to visualize the complete
generic development pathway in a concentrated rational picture.
Review the checklist titled Initial Regulatory Check Prior to Pre-formulation in order to obtain
an initial overview of the forensic regulatory elements.
An early decision on pack type and sizes is required from the marketing and sales
department in order to initiate the product's stability specifications.
Part Two
Part Two of the Handbook of Pharmaceutical Generic Development Series provides
a full scale development of a current US abbreviated new drug application (ANDA)
for the selected dosage form chosen (e.g. tablets, capsules, semisolids, liquids etc.)
In the development ANDA, pages and sections provide an example of each and
every document required to compile and assemble a complete regulatory file for
submission to the FDA’s Office of Generic Drugs (OGD).
The left hand side pages are designed for personal notes, summaries and
explanations on the submitted data forms and tables. These pages are designed for
side-by-side comparisons and ease of viewing. Essential tips and potential traps are
explained in the notes and summaries as well as the common do’s and don’ts
regularly addressed by the agency review chemists in a ANDA / AADA file
submission.
SUMMARY:
◊ Review the ANDA requirements and the relevant guides and guidelines
impacted by the dosage form with the Regulatory Affairs Department
◊ Know all the twenty two (22) ANDA sections in the registration file impacting on
the development, formulation, manufacturing procedures, all intermediate and
final product specifications, pivotal batch, stability requirements, as well as key
regulatory ANDA do’s and don’ts.
◊ Complete the pre-formulation regulatory checklist and discuss and review all
the paperwork required to complete this section successfully.
SECTION I
Section One of the ANDA file consists of three documents. A drug developer needs
to understand the information required to accurately compile these three documents
as the information impacts upon the development timeline. These three parts are:.
♦ Cover Letter and Field Letter
♦ Table of Contents
♦ Signed Application Form
Cover Letter
The cover letter should be on the letterhead of the Applicant or the Applicant's Agent
and should state the following particulars:
Table of Contents
Table of Contents laid-out to the FDA's CDER Guide to Industry Format, (Feb. 1999
Overall ANDA Guideline Requirements) needs to provide rapid access to the twenty
Two (22) sections of the ANDA submission file. A drug developer needs to
understand all 22 sections, in detail, as they contain the overall development,
manufacturing and control data that makes up the entire drug development project.
Careful review of each section and its required contents will aid significantly in the
pre-formulation to final pivotal processes. The overall development report can be
structured on each CMC section highlighting the choices made and work carried out
to fulfill each sectional requirement. The development report is a real-time ongoing
series of procedural reports that culminates in the final collated Development Report.
SIGNED APPLICATION FORM:
Signed Application Form (Form FDA 356h or 3439) with original ink signature
requires all DMF numbers of materials and containers used in the final product
presentation. Also no test results, analysis, or stability results may be submitted from
a QC / QA or R&D laboratory that has not received a FDA Plant DMF number (Note:
- New Revised Form FDA 356h became official from January 8, 1998).
Obtaining DMF numbers from suppliers on time requires advanced pre-planning (up
to three months or more) and scrupulous care should be taken that no Agency
deficiencies or FDA concerns of a recent or overdue nature are outstanding on the
materials or container components being used in the firm's generic formulation.
ØCHECK LIST×
CL # HPGD-03-01Y2K
S
first
tandard operational procedures for
a generic development unit are the
essential documentation
information and attached to the SOP.
Don’t - have loose lying SOPs
gathering dust on the shelf.
requirements.
Without a functional set of standard Don’t - write a SOP that can not be
development procedures, developing followed - routinely, by all
generic drugs will follow a haphazard concerned.
non-reproducible process. SOPs are Don’t - write long SOPs (about 1 to
efficient and useful instructional and 3 pages is sufficient for the average
working tools. SOP).
Standard Operating Procedures Don’t - write SOPs just to comply
should meet six basic and functional with GMP - use them frequently as
requirements operational and training tools.
Ø CHECK LIST ×
CL # HPGD-02-01Y2K
SOP CONTROL.
1. Is there an Index for all the Pharmaceutical Development SOPs ? qYes qNo
2. Is this Index a Standard Operational Procedure itself ? qYes qNo
3. Are SOPs correctly organized according to this Index ? qYes qNo
4. Are the SOPs available in the community language(s) ? qYes qNo
5. Do the SOPs have a logical, intelligent SOP numbering system ? qYes qNo
6. Are all SOPs signed and is each signature dated ? qYes qNo
7. Check, if the circulation of SOPs is effective and on-time ? qYes qNo
8. Check that no SOP has a date older than two (2) years ? qYes qNo
9. Check that superseded SOP editions have all been removed ? qYes qNo
10. Check that all department personnel have signed the ‘Agreement to qYes qNo
Comply with SOPs’?
11. Is the effective date of the SOP about 21 days after the last qYes qNo
signature - to allow for SOP training ?
12. Has the SOP been signed by all authorizing parties within 14-21 qYes qNo
days of the first signature?
13. Are the department SOPs - brief, concise and to the point? (select qYes qNo
and review three examples.)
14. Are the ‘responsible persons’ clearly indicated in the SOP's qYes qNo
Responsibility section?
15. Has a product specific ‘Development SOP’ been prepared for each qYes qNo
dosage form under development (i.e. SOP for Tablets IR and Tablets
CR/MR etc.)?
16. Do these ‘Development SOPs’ acts as a blue print for the qYes qNo
development team to follow to ensure complete product development?
17. Does the development team regard ‘Development SOPs’ as qYes qNo
valuable working tools and fail-safe procedural mechanisms?
Footnote : Bold lettering in checklist indicate that this work must be checked and approved before pre-formulation work starts.
ØCHECK LIST×
CL # HPGD-02-01Y2K
DEVELOPMENT NOTEBOOKS.
‘…Sign every stage not every page - check every page and every stage….’
1. Does the R&D unit have bound & page numbered notebooks ? qYes qNo
2. Are the development notebooks signed on every stage & page ? qYes qNo
3. Has each completed section been signed with a check signature ? qYes qNo
6. Are all pre-formulation and development formula numerically recorded ? qYes qNo
7. Do the notebooks record successful and failed development product formula ? qYes qNo
8. Do all calculations have check signatures which are dated. qYes qNo
9. Has the Product Development SOP been complied with during the product qYes qNo
development phase ?
10. Is a process of formula, specifications and process optimization evident ? qYes qNo
11. Do all specifications have an appropriate range, where absolutely needed? qYes qNo
12. Have critical upper and lower range limits been qualified ? qYes qNo
14. Are the lot #’s, expiration dates and source of each active and non-active qYes qNo
ingredient used during routinely recorded in workbooks ?
15. Does the development notebook appear suitable as a scientific basis for the qYes qNo
generic drug product ‘Product Development Report’?
Footnote : Bold numbers in checklist indicate that this work must be checked and approved before pre-formulation work starts.
STANDARD OPERATING
PROCEDURE
SOP # HPGD-02-01Y2K
SOP CONTROL
# HPGD-02-01Y2K Indexing procedure for pharmaceutical development SOPs
# HPGD-02-01Y2K Index for pharmaceutical development SOPs
# HPGD-02-01Y2K Signing procedures of pharmaceutical development SOPs
# HPGD-02-01Y2K Standard Operating Procedures - number and format
# HPGD-02-01Y2K Circulation of pharmaceutical development SOPs
# HPGD-02-01Y2K Annual review of pharmaceutical development SOPs
# HPGD-02-01Y2K List of FDA guides and Guidelines impacting on product
development dosage form and type (IR / CR)
DEVELOPMENT NOTEBOOKS
# HPGD-02-01Y2K Issue and use of pharmaceutical development notebooks.
# HPGD-02-01Y2K Signing procedures for development notebooks.
# HPGD-02-01Y2K Development notebooks - review and audit procedures.
# HPGD-02-01Y2K Standard procedures in generic product development.
# HPGD-02-01Y2K Pharmaceutical development notebooks - disposition.
AN OVERVIEW
ØC H E C K L I S T ×
CL # HPGD-03-01Y2K
1. Is a ‘Development SOP’ for the specific dosage form available? qYes qNo
2. Does a SOP specifying the contents of a 'Product Development Report'? qYes qNo
3. Is the active ingredient characterized for particle size and bulk density for each qYes qNo
approved supplier (a least two suppliers should be evaluated and approved)?
4. Are the source and supply of the excipients characterized and listed? qYes qNo
5. Is the source and supply of the complete container-closure-system characterized qYes qNo
6. Does the SOP indicate that non-compendial active material assays requires a qYes qNo
validation and stability indicating assay?
7. Does the SOP required full analysis of the Reference Listed Drug (RLD), its qYes qNo
overall impurity and stability profile?
8. Is an historical listing and summary of all experimental batches manufactured qYes qNo
required?
9. Is a multipoint dissolution profile of the product formula at key stages required qYes qNo
and compared to the RLD?
10. Do the manufacturing procedures, process parameters and in-process controls qYes qNo
require optimization and challenges at either ends of the specification's limits?
11. Is a process of tightening and qualifying the product specifications, (based on qYes qNo
batch analysis) evident, as the development process undergoes optimization?
12. Does the development process identify the critical processing steps, for the qYes qNo
process validation protocol, with the potential to affect the product?
13. Are hardness vs. dissolution tests qualified (Tablet Hardness Qualification)? qYes qNo
14. Does the analytical development require a final validated assay and stability qYes qNo
indicating (SI) assay well before running the PQ and Pivotal batch?
15. Are stability study assays of the PO and PQ batch required to be tested by a qYes qNo
validated assay procedure and stability indicating (SI) analysis?
Footnote : Bold numbers in checklist indicate that this work must be checked and approved before formulation work starts.
Formula Development
‘…research and evaluate the reference listed drug thoroughly…'
ANDA Tablet Preparations Non-active ingredients (excipients)
The development of a single dosage The product formula of the generic
oral tablet requires six key decisions. single dose oral tablet do not have to
This evolves choosing the : - contain the same inactive ingredients
at the same quantities as the RLD as
♦ Reference listed drug (RLD) sterile or semi-solids dosage forms.
♦ Active material (source/supply) The qualitative ingredients are
♦ Non-active ingredients (source/supply) required to be in the OGD Inactive
Ingredient Guide (IIG).
♦ Container-closure system (as RLD) The FDA publish the Inactive
♦ Comparative dissolution procedure Ingredient Guide. Inactive ingredients
♦ Bioequivalence to the (RLD) found in approved oral tablets drug
The RLD is chosen from the FDA products in the US are listed in the IIG.
'Orange Guide' (now on the Internet). The strength (i.e. quantity) of the
(‘Approved Drug Products with Therapeutic inactive ingredient in the product
Equivalence Evaluations’ - Ed. 20th,2000) formula must not be greater than the
Active substance highest concentration previously
The active drug substance is chosen approved in an approved drug product
according to standard criteria. Correct for the same route of administration
choice of active ingredient is critical (i.e. oral route)
and time consuming. Key choosing Several SOP’s have been included to
parameters include :- clarify the choice of inactive
♦ Analytical profile - similar to RLB ingredients for pre-formulation and
under normal and stress conditions. formulation development.
♦ Impurity profile - similar to RLB The choice of a well known excipient
under normal and stress conditions. manufactures with an established
excipient range is very important as
♦ Approved supplier must meet all long term stability, dissolution and
ANDA regulatory documentation aging problems are minimized or
requirements. avoided. Thus source and supply of
♦ Active Material specifications inactive ingredients is paramount.
remain constant - batch-to-batch
Container-closure-systems
(not a R&D lot or non-commercial
The drug product container-closure
batch)
system should be a similar material
♦ Vendor able to supply for the next 8- composition as the RLD container-
10 years at similar specifications to closure system.
material used in actual pivotal The degree of product protection by
batch. the container-liner-closure system
Using the RLD’s Active Material must prevent physical, chemical and
Ideally the same supplier of the active microbiological changes on storage
drug substance as used by the RLD, is and during customer-consumer use.
the most cost-effective in the long (FDA final guidance for industry on container
closure is dated May 1999.)
term, as stability, impurity, and aging
profiles are similar.
ØC H E C K L I S T ×
CL # HPGD-02-01Y2K
FORMULA DEVELOPMENT
‘…Systematically compare your developing product to the chosen RLD
at all key stages…'
1. Has the Reference Listed Drug (RLD) been chosen from the Orange qYes qNo
Guide?
2. Has the RLD been purchased in all the proposed marketing sizes ? qYes qNo
3. Have different batch numbers (3 lot #’s) of the RLD been purchased? qYes qNo
4. Confirm if the RLD is of recent manufacture (analyze new samples)? qYes qNo
5. Conform that at least 10-20 samples of each RLD lot # and pack size qYes qNo
are available for physical, chemical (assay and impurities), dissolution
and stability testing?
6. Confirm if the RLD has been placed on stability at 40o C for 3 months qYes qNo
for evaluating potential degradation and impurity levels?
7. Confirm if the impurity profile of the RLD has been evaluated? qYes qNo
8. Has reverse engineering of the RLD formula been performed? qYes qNo
9. Have the chosen inactive and maximum strength been cross-checked qYes qNo
in the IIG? (for unique or unusual excipients)?
10. Are the in-actives qualitatively compatible with the RLD for oral use qYes qNo
(composition and strength)?
11 Have the RLD formula been reviewed in the International Drug qYes qNo
Compendia (Italian, French, Swiss) for formula composition data?
12. Has the FOI system been used to gather data on the Innovative qYes qNo
drug?
13. Has a full analytical profile range been determined from analysis of qYes qNo
the various batch lots of the RLD (at least 3 lots #’s for Assay; Content
Uniformity; Impurities; Dissolution)?
14. Has the chosen RLD undergone stress testing to establish the level qYes qNo
of its degradation products?
15. Has a multipoint dissolution of the several RLD batch lots been qYes qNo
evaluated to assess the consistency of the RLD dissolution parameters?
Footnote : Bold numbers in checklist indicate that this work must be checked and approved before formulation work starts.
PROCEDURES
SOP # HPGD-02-01Y2K
FORMULA DEVELOPMENT
The following selected model Standard Operating Procedures are recommended for
a generic development unit : Models of selected SOPs are provided.
DEVELOPMENT SOP
HPGD-02-01Y2K Setting up a General Development SOP.
HPGD-02-01Y2K Setting up a Product Specific Development SOP.
HPGD-02-01Y2K Contents of a Development SOP - (Oral Tablets)
DEVELOPMENT FORMULA
HPGD-02-01Y2K Vendor Certification Requirements for Product Development.
HPGD-02-01Y2K Formulation of ANDA (Oral Tablets) Preparations
HPGD-02-01Y2K Formulation of ANDA to Q & Q Status (Oral Tablets)
HPGD-02-01Y2K Standard Procedures for Generic Product Development
DEVELOPMENT REPORT
HPGD-02-01Y2K SOP for Development Reports.
HPGD-02-Y2K Contents of a Development Report - (Oral Tablets) - Single
dose.
HPGD-02-01Y2K Parameters for Process Optimization and Process
Qualification.
NOTE ON DEVELOPMENT
The intent and purpose of the pivotal batch is as a final demonstration that the
formula, process and controls are well developed and tested during development
stages and really need no significant changes or further process qualification.
However scale-up changes can take place within the SUPAC rules after
manufacturing the pivotal batch.
These SUPAC rules govern the Scale-Up from pivotal (10% or more) to commercial
(100%) and Post-Approval Changes i.e. changes after registration approval has
been given.
[End of Document]
STANDARD OPERATING
PROCEDURES
SOP # HPGD-02-01Y2K Page 1 of 1
Biopharmaceutical Guidance
FDA Web site Listing of
In Vivo Bioequivalence and In Vitro Dissolution Testing
1. Alprazolam Tablets 19. Hydroxychloroquine Sulfate Tablets
APPROVE
STAGE 1 a minimum of
LITERATURE TWO
SEARCH SUPPLIERS
STAGE 2
ACTIVE SOURCING
Q3A
Impurities cf.
STAGE 3 innovator's profile
Do not evaluate ACTIVE EVALUATION
material while still
in a R&D
STAGE 4
stage.
ACTIVE PURCHASING
USE ONLY
PRODUCTION
ACTIVES
STAGE 5
Active testing
STAGE 6
Innovator Product Purchasing
Purchase a new
STAGE 7
lot number every 3 months
Innovator Product Testing
from the smallest to the
largest pack size
(in each dosage strength)
STAGE 8
Bulk Active Testing
DEVELOPMENT
21 STAGES STAGE 10
Container Closure
System Choices
STAGE 11
Manufacturing Process Evaluation
STAGE 12
Bulk Active Purchase
PRODUCT DEVELOPMENT
FLOWCHART
Solids Dosage Forms
STAGE 13
Analytical Evaluation
STAGE 14
Process Optimization
PO Batch
STAGE 15.
Prepare full Written SCALE - UP
Protocols for PO
Scale-Up & PQ STAGE 16
Batches PROCESS
(Future Q6A
QUALIFICATION
Requirements will impact
on this development)
STAGE 17
PIVOTAL BATCH
PRODUCTION
STAGE 18
ANDA PRE-SUBMISSION
AUDIT
Purchase (Potential) Evaluate at least two potential active material suppliers for
Active Material approved supplier status
Stage 5 Active Testing
Testing of Active Chemical testing by the R&D analytical lab as per
Material sample a. Pharmacopoeia monograph (if present)
b. Pharmacopoeia Forum (if available)
c. In-house method (based on manufacturer)
d. Supplier's test methods and specifications
PRE-FORMULATION
Development Scope of Product Development
Stage
Stage 6 Innovator's Product Purchasing
DRUG PRODUCT Purchase at least 3 different lots in smallest and largest pack size
Innovator Samples for each product strength
DEVELOPMENT BATCHES
Development Stage Scope of Product Development
Stage 9 Excipients
Evaluation of formu-
Excipient compatibility using DSC methods and stability
lation with suitable
assessment
excipients
Stage 10 Container Closure System
Evaluation of suitable Choice of container-closure-liner system including:
Container-Closure • material composition,
System • type of thermoplastic resin and resin pigments,
• manufacturers and suppliers,
• liners and seals used by closure manufacturer,
• cotton and desiccants.
• manufacturer's DMF numbers for all component parts
• Letters of Access for regulatory authorities to view DMF dossiers
Stage 11 Manufacturing Process
PROCESS OPTIMIZATION
Development Scope of Product Development
Stage 14 Process Optimization
GRANULATION • Effect of granulation parameters
OPTIMIZATION
• Granulation time
• Speed of choppers (I & II) or mixer blades
• Solvent addition rate and overall amount
• Ratio of intra-granulate Disintegrant and binders agents
• Screen size for milling (e.g. 0.6 or 0.8mm)
• Adjusting mill screen size up or down to fine tune hardness
• Evaluation of optimized granulate and tablet attributes
DRYING • FB Drying temperature versus target LOD and range limits and
the effect on granulate and tablet properties (flow, capping,
sticking).
SCALE UP
Development Scope of Product Development
Stage 16 SCALE UP
Scale-up Scale-up lot prepared if larger batch size scale up problems
anticipated.
Process Qualification batch and Scale-up batch may be
evaluated as a single batch.
Scale-up Report The preparation of a Scale-up Report. The Scale-up report forms
part of the overall Development Report
PROCESS QUALIFICATION
Development Scope of Product Development
Stage
Stage 17 Process Qualification
The process qualification batch is manufactured in order to detect any problems that may arise during
the manufacture of production size batches, allowing a solution prior the manufacture of the pivotal
demonstration batch. Scale-up to the pivotal batch size or 70% of the pivotal batch may be combined
with qualifying the manufacturing process At this stage full manufacturing documentation is prepared
alone standard procedures.
PRODUCTION Process Qualification batch should be compressed in a production
FACILITIES (or production type with same principle and operation) tabletting
machine
Size of pivotal and marketing batch confirmed (NLT 100 000 net/
packed at target parameters or 10% of proposed market batch).
PROCESS QUALIFICATION
Development Stage Scope of Product Development
Stage 17 (Cont) Process Qualification
FINAL REVIEW and Review of proposed formula, manufacturing process and control
AUTHORIZATION parameters with production personnel and QA Staff with
authorization signatures (RD; QA-QC; RA; and Production)
KEY STEPS Critical manufacturing steps designated and sampling and testing
parameters specified.
P.Q. REPORT Upon completion prepare Process Qualification Report. This P-Q
report forms part of the overall Development Report
PIVOTAL BATCH
Development Scope of Product Development
BIOEQUIVALENT STUDY
Stage Scope of Product Development
Stage 19 BIOSTUDY Evaluation
BIOSTUDY Fasted Perform Fasted / Food Effect Biostudy on Pivotal Lot Samples
BIOSTUDY Perform Food Effect Biostudy on Pivotal Lot Samples (See food
[Food Effect] effect guidelines, where appropriate)
HIGHEST DOSAGE Biostudy generally performed on highest strength of product
One or two studies Fasted AND Food Effect Study may be required
WAIVER For multiple strength products Invitro dissolution testing conducted
CONDITIONS in three different pH media on lower dosage forms
SIMILARITY TESTING Perform Similarity Test [F2 Test] on dissolution results.
PRE-SUBMISSION AUDITING
Development Stage Scope of Product Development
ANDA Regulatory File Audit Plant and Laboratory Documentation as per ANDA
Biostudy Report Evaluate and develop a IVIV correlation (Level A where possible.)
ANDA SUBMISSION
Development Stage Scope of Product Development
Stage 21 ANDA Submission
(1 Field Copy)
VALIDATION BATCHES
Development Scope of Product Development
Stage
COMMERCIAL RE-VALIDATION
DUE TO MAJOR CHANGE
Development Scope of Product Development
Stage
Formula Change Revalidate procedure with new formula process or equipment with
Equipment Change
The invitro dissolution settings are adjusted (via media, pH agitation) until a I : I
correlation is achieved (Level A) or a single dissolution point and a plasma
parameter is shown to correlate (Level C).
When more than one point correlates a multiple Level C is obtained - which may
possibly be upgraded to a Level A with additional development work.
This matching of dissolution settings with plasma levels, that are derived from a
specific IR formula and its corresponding manufacturing process, is in fact simply an
arbitrary set of values that establish the so called 'predictive mathematical model'.
DC TABLET DEVELOPMENT
D irect Compression
Tablet Development
‘…Developing low strength actives via direct compression
is often the preferred route…
DIRECT COMPRESSION
Development Flowchart
EVALUATE
Weight uniformity
Content Uniformity
FORMULATE Hardness & Dissolution
Optimise Development Batches Profile
Dry blend mix A, B & C (compare active dissolution)
+ compare reference drug)
FINE TUNE
OPTIMIZATION A, B & C Repeat tests
Unlubricated blend - UoC Adjust D & E
lubricated blend - UoC
Lubrication range (0.5 - 1.0%)
Tablet Weight Uniformity
OPTIMIZATION
Formula & Process
Fine tune D & E
Tablet Hardness
Qualification (low & High) To Pivotal
with Dissolution Profile PROCESS QUALIFICATION
DIRECT COMPRESSION
SUITABILITY:
Glibenclamide 5 mg
Selegiline 5 mg
Manufacturing Flowchart [I] Famotidine 10/20/40 mg
Bromhexine 8 mg
SDL(30%) - MCC(60%) Amitriphyline 10 mg
LACTOSE
Active Lubricant
Material and Glidant
MIX Y-cone
SIEVE
Mill FLUSH
50 MESH
Where necessary
0.6 -0.8 mm
MIX
MICROCRYSTALLINE
CELLULOSE rpm + time
SUPER
DISINTEGRANTS Lubricant coats
the lactose
MIX Y-cone providing
greater control
over final
blending stage
Development
Content Uniformity on
unlubricated blend to
establish mixing time
COMPRESS QC
Weight uniformity
Thickness
Hardness
AQUEOUS Roller mixer Disintegration
FILM COAT
RELEASE
ID
To printing COATED DC TABLETS Assay
Content Uniformity
Dissolution
DIRECT COMPRESSION
Manufacturing Flowchart [II]
SDL(60%) - MCC(30%)
Active LACTOSE
(65%)
Material (10%)
MICROCRYSTALLINE
35% CELLULOSE (15%)
Lubricant
(1%)
SODIUM STARCH
GLYCOLATE (2%)
SIEVE
AEROSIL (0.5%)
STARCH (6%)
20 - 25 min
Aerosil mixed with either
starch (4), lactose (1+3)
Sieve
or lubricant (2)
(Where necessary)
0.8 mm Lubricant coats
final blend
in Y-Cone
directly for a
MIX Y-cone
short period
(3-5 min)
20 - 25 min
During
Development
Content Uniformity of
unlubricated blend to
establish mixing time
BLEND Y-cone IPQC
5 min ID
Assay
Content Uniformity
COMPRESS QC
Weight uniformity
Thickness
Hardness
AQUEOUS Roller mixer Disintegration
FILM COAT (See Chapter 11)
RELEASE
ID
To printing COATED DC TABLETS Assay
Content Uniformity
Dissolution
STRENGTH Per TABLETÜ 1mg 2mg 5mg 5mg 8mg 10mg 5mg/ 10mg
10mg
ACTIVE Ü [A] [B] [C] [D] [E] [F] [G] [H]
% Active Material 0.7 1.4 3.6 5.0 5.5 10.0 3.55 5.0
Magnesium Stearate 0.5 0.5 0.6 0.5 1.2 1.0 0.65 0.5
TOTAL PERCENTAGE 100 100 100 100 100 100 100 100
Tablet weight (mg) 140 140 140 100 150 100 140/ 200
280
Disintegration <<15 MIN <5 <5 <5 <5 <5 <5 <5 <5
[A] Flunitrazepam DC Tablet [D] Selegiline DC Tablet [G]Buspirone DCTabs.
[B] Flunitrazepam DC Tablet [E] Bromhexine DC Tablet [H]Ketorolac DC Tabs.
[C] Glibenclamide DC Tablet [F] Amitriphyline DC Tablet
STRENGTH Per TABLETÜ 1mg 2mg 5mg 5mg 8mg 10mg 5mg 10mg
10mg
ACTIVE Ü [A] [B] [C] [D] [E] [F] [G] [H]
% Active Material 0.7 1.4 3.6 5.0 5.5 10.0 3.55 5.0
TABLET DEVELOPMENT
W et Granulation Development
OverView
Tablet Development
‘…Developing low & high strength actives via wet granulation
high shear techniques…
Table 2.
ALCOHOL / WATER GRANULATION
Wet Granulation Design Occasionally an appropriate ratio of
Parameter Decision Range alcohol / water mixtures from 60 : 40
Wet granulation Use a standardise (Nabumetone) to 95 : 5 (Mesalamine,
Excipient blend excipients range ISMN etc.) are appropriate and can
Multiple strength geometric design produce stable granulated material.
Tablet Weight 150 - 980 mg Table 4.
Individual weight ± 7.5% Alcoholic Granulation
Average weight ± 5.0% Active Alcohol/Water
Tablet Hardness 8-18 SCU
Nabumetone 500/750 60.0 : 40.0%
Thickness 2.5 - 8.0mm
Mesalamine 95.0 : 5.0%
Friability < 1.0%
Disintegration <10 -15 min AQUEOUS GRANULATION ACTIVES
Dissolution NLT 80 in 45 min Table 5.
Aqueous Granulation
CHOOSING THE FORMULA
Active % P. Water
n Choice of alcoholic (95% or 99.5%)
or aqueous granulation procedure. Carbidopa /Levodopa 100
The choice of the granulating solvent Clonazepam 100
whether pure alcohol or a water / Enalapril Maleate 100
alcohol mix is a critical early decision. Famotidine 100
ALCOHOLIC GRANULATION Frusemide 100
Many active materials do not produce Gemfibrozil 100
satisfactory stability profiles when Norfloxacin 100
granulated solely with purified water. Tamoxifen 100
Active degradation and overall Trazodone HCl 100
impurity growth is often accelerated
during wet granulation or during the n CHOICE of the wet granulation
shelf life period due to the presence of excipient mixture.
residual water in the compressed The percentage of the active
tablet. Hydrous (95%) or anhydrous ingredient in the overall formula may
alcohol (99.5%) is the granulating range from 0.5 mg (Clonazepam) to
solvent of choice in aqueous sensitive 850 mg (metformin) while excipient
active materials. Table 3. composition may vary from 99.5% to
less than 10% (see tables 15 to 17).
Alcoholic Granulation Where the active material exceeds
Active Alcohol % 90% with 10% or less of excipients the
Bupropion 99.5% process of choice often rests in
Carbamazepine 95.0% fluidized bed granulators / processors.
Diclofenac Na / K 99.5% (ref. Table 16).
Glipizide 95.0%
The active material's particle size and
ISMN 40mg 95.0%
bulk / tapped density are significant
Mesalamine 95.0%
parameters in wet formulation.
Naproxen 95.0%
Processing by high shear granulators
Omeprazole 99.5%
produce high density granules as
Simvastitin 95.0%
opposed to moderate density granules
Verapamil HCl 99.5%
produced in fluidized bed granulators.
Alternatively after spraying and drying The ideal granulate should be readily
the 'granulated active' is transferred to compressible with superior flow
a Y-cone or tumble blender and the properties at high compression
remaining 50% disintegrant blended. speeds. Disintegration should be rapid
The final blending step requires the (5-15 minutes) at optimum target
addition of the lubricant and a brief hardness values that may range
blending (<5 min/10 rpm). Blending in between 8-18 SCU. The dissolution
a Y-cone via a 'two-stage blending profile should be designed to be just
process' is a common practice. marginally greater than the solubility
Table 6
of the pure active material under
Spray Dried Actives similar dissolution conditions.
Active % Excipients
Tablet/caplet thickness and
Material mg per tablet
dimensions depend on tablet weight
Famotidine 750 Strong Binder 2-5 and die chosen to meet a thickness of
Metformin 850 Super Disintegrant ±4
between ±2.5 and 8.0 mm.
Lubricant ± 0.8
ADVANTAGES:
Ü Single or dual Lubricant
Fluidized Bed Processors have the Regular lactose, tricalcium Phosphate
following properties. and the various dried starches are
Ü Capable of producing granules with common diluting and bulking agents
90-93% active and 7-10% non-actives and perform well in wet mixing and dry
Ü Narrow particle size distribution blending operations in amounts from
Ü Granulation end-points may be 2-20% (used in lower cost drug
fixed by controlled spraying formulations).
DISADVANTAGES: For high shear granulators, 200 mesh
Ü Process a narrower range of lactose is preferred due to its superior
starting materials flow and granulating properties. Table
Ü Produces moderate density 7 shows common diluents used in
granules aqueous and alcoholic granulation.
Ü Problems with low density or high Dried starch NF (DS), pregelatinised
density actives and excipients starch (STA-RX-1500) and sodium
starch glycolate are frequently used.
Table 7.
Avicel pH 101 / 102 /105 /112 when
Bulking Excipients blended in approximately equal parts
Non-active % / tablet 50 : 50% intra and extra-granularly,
Microcrystalline Cellulose 10 - 30 form compressible granules with
101/105 /102/112 superior disintegration abilities (when
Lactose MH-200 35 - 75 used at quantities that range from 15,
to 30, or 60%.
Tricalcium Phosphate 10 - 50
Equivalent results are obtainable by
Dried starch 5 - 25 substituting a 1:1 ratio (equal parts) of
Maltose - Advantose™100 35 - 75 crystalline maltose and micro-
crystalline cellulose.
For 'fine particle-size actives', 200 Table 10.
mesh lactose monohytrate is more Granulating Agents / Binders
suitable. With 'granular actives' the Excipient % per tablet
100 mesh lactose monohytrate is
Povidone K30 2-4
generally preferred during the
blending stage. Povidone K90 2 - 3.5
Microcrystalline lactilol (a Hydroxypropyl Cellulose 1 - 1.5 - 2.5
disaccharide sugar alcohol derived Hydroxypropyl 1 - 1.5 - 2.5
from lactose) may be substituted in Methylcellulose
lieu of lactose. Marginally harder Pregelatinized Starch 1 - 5 - 10
tablets are produced at the same (Starch 1551™)
compression strength. Since
microcrystalline lactilol has a lower Where necessary suitable additional
moisture pick up and lower disintegrants such as sodium starch
hygroscopicity, shelf life extension is glycolate (2-5%) or 'dried' starch NF
promoted when used with active (~3% to 6%) promote rapid tablet
materials sensitive to moisture pick-up disintegration and allows faster drug
over the shelf life testing period. release. Where Mannitol formulations
0rganic acids may significantly tend to increase the dissolution time
improve the overall stability profile of on ageing (shelf life period), sodium
certain actives (e.g. Bupropion HCl). starch glycolate (SSG) is
Table 8. recommended (at about 3.0 - 5.0%).
pH adjusters /Organic Acids Table 11
Non-active % / tablet Extragranular Disintegrants
Citric Acid 2-4
Excipient % per tablet
Maleic Acid 1-2
Sodium starch glycolate 2.0 - 4.0
Glutamic Acid HCl 10 - 20 Croscarmellose Na 3.0 - 5.0
Avicel pH101™ - microcrystalline cellulose (MCC) Starch NF (dried) 3.0 - 5.0
Advantose™ - crystalline maltose (CM)
Microcrystalline 5.0 - 15.0
Table 9. Cellulose 102
Super Disintegrants Microcrystalline 5.0 - 10.0
Non-active % / tablet Cellulose 105
Croscarmellose Na 2-5 Common splitting ratios between intragranular:
extragranular disintegrants are in the order of 1:1
Crospovidone 2-4 and 1.5 : 1
DRYING
FLOW Range from Stage
CHART 1 1.5 - 4.5%
Blend
Uniformity testing
COMPRESS
COAT (Target speed Rpm)
Flow Chart 2
WET GRANULATION
Development Flowchart
EXCIPIENT MIX - DECISION TABLE
A B C D E
Filler Intragranular Granulating Agent Extra disintegrant Fine Tuning
Disintegrant (Fine Tuning Glidant / lubrication
Dissolution)
e.g. L/MCC MCC/SSG / CC PVP/HPMC/HPC SSG / CC / XP Talc / SA /
CSD/MS
KEY
CC Croscarmellose
CSD Colloidal Silicon Dioxide
L Lactose monohydrate Evaluate Active material
MS Magnesium stearate Dissolution ; Bulk Density ; Particle size, EXAMPLE:
MCC Microcrystalline cellulose
% Active in formula. 22% SRx
SA Stearic Acid Wet
PVP Povidone K30 /K90 35% MCC
HPC Hydroxypropyl cellulose 1.0% HPC
S Starch NF 11% S
SRx Pregelatinized Starch
10% MCC
SSG Sodium starch glycolate CHOOSE GRANULE EXCIPIENTS Dry 0.8 CSD
XP Crospovidone
Select A, B, C, D & E 1.0% MS
from table above & refer to tables 15-17
Fine Tune
MCC type pH 101/102/105
Disintegrant - (SSG/MCC) Flat, bevelled or
Lubricant package CHOOSE Biconvex
Based on the active Tablet Weight & Shape Choose higher
material's properties ± 200 for 1-10% Active or lower % lubricant
± 550 for 50% Active
± 750 for 80% Active
EVALUATE
Weight uniformity
Content Uniformity
FORMULATE Hardness & Dissolution
Optimise Development Batches Profile
blended mix A, B, C, D & E (compare active dissolution)
+ compare reference drug)
FINE TUNE
OPTIMIZATION A, B & C Repeat tests
Unlubricated blend - UoC Adjust D & E
lubricated blend - UoC
Lubrication range (0.5 - 1.0%)
Tablet Weight Uniformity
OPTIMIZATION
Formula & Process
Fine tune D & E
To Pivotal Process
Tablet Hardness
Qualification (low & High)
with Dissolution Profile
PROCESS QUALIFICATION
Flow Chart 3
WET GRANULATION
Manufacturing Flowchart [I]
LMH
and/or
MCC
SIEVE
DRYING FLUSH
50 MESH
LOD CHECK
Mill MIX
Where necessary
0.6 -0.8 mm
rpm + time
EXTRAGRANULAR
MCC and / or SSG
Lubricant coats
glidant
SUPER
providing
DISINTEGRANTS
greater control
MIX Y-cone over final
blending stage
rpm (10) + time (20 min)
During
Development
Content Uniformity on BLEND STEP IPQC
unlubricated blend to ID
establish mixing time rpm (10) + time (5 min)
Assay
Content Uniformity
QC
COMPRESS Weight uniformity
Thickness
Hardness
Friability
AQUEOUS Roller mixer Disintegration
FILM COAT
RELEASE
ID
To printing COATED TABLETS Assay
Content Uniformity
Dissolution
Flow Chart 4
WET GRANULATION
Manufacturing Flowchart [2]
ALCOHOLIC - (Product Specific Chart)
SIEVE
50 MESH
During GRANULATOR
Development 3 - 5 min
Establish
Content Uniformity of
unlubricated blend to DRY
fix the mixing time
LOD CHECK
Lubricant coats
Sieve final blend
Aerosil mixed with (Where necessary) in Y-Cone
either lubricant or 0.6 - 0.8 - 1.0 mm directly for a
starch short period
MIXING (3-5 min)
Tumbler/Y-cone
AEROSIL (0.5%)
STARCH (6%)
MICROCRYSTALLINE 20 - 25 min
CELLULOSE (18%)
IPQC
BLEND Y-cone ID
rpm (10) + time (5 min) Assay
Content Uniformity
SUITABILITY: QC
Diclofenac Na 50 mg
Weight uniformity
COMPRESS Thickness
Hardness
Friability
AQUEOUS Roller mixer Disintegration
FILM COAT
RELEASE
ID
To printing COATED TABLETS Assay
Content Uniformity
Dissolution
ÜACTIVE MATERIAL %Ü 50.8 1.14 10.0 5.0 10.0 27.3 58.8 10.9*
%
Hydroxypropyl Cellulose NF - - - 1.0 1.0 - - -
Color/Dye 3.5 1.4 0.2 - - 0.1 - -
Sodium Bicarbonate USP - - 1.5 - - - - -
Aerosil - - - 0.8 0.8 0.8 0.5 0.5
(Colloidal Silicon Dioxide NF)
Magnesium Stearate 0.5 0.6 0.5 1.0 1.0 1.0 0.8 1.0
Purified Water USP qs qs qs qs qs qs qs qs
TOTAL PERCENTAGE Ü 100 100 100 100 100 100 100 100
Tablet weight (mg) Ü 250 180 200 210 210 150 680 280
Hardness SCU (target) 9 8 8 7-9 9 9 10 9-11
Disintegration <15 MIN 5-15 5-15 5-15 5-15 5-15 5-15 5-15 5-15
KEY:-
[A] Carbidopa/Levodopa [B] Clonazepam [C] Enalapril Maleate [D] Famotidine USP
[E] Famotidine USP [F] Frusemide BP [G] Norfloxacin USP [H] Tamoxifen Citrate*
STRENGTH Per TABLETÜ 275 40 150 400 450 250 500 200
mg mg mg mg mg mg mg mg
ACTIVE Ü [I] [J] [K] [L] [M] [N] [O] [P]
Ü ACTIVE MATERIAL %Ü 72.0 36.0 37.5 59.7 69.0 90.9 90.9 52.6
Avicel pH 101 - - - - - - - -
Avicel pH 102/105 7.0 - - 18.3 15 - - -
Lactose monohydrate 100 mesh - - - - - - - -
Processed in
Lactose monohydrate - 36.0 49.0 - - -
Fluidized Bed 36.6
200 mesh NF Granulator
Lactose Spray Dried NF - - - - - - - -
Sodium Starch Glycolate - - 10.0 - - 6.0 6.0 -
Croscarmellose Sodium 2.0
Starch STA-RX-1500 - - - - 8.5 - - 5.0
Starch NF - 25.0 - 4.2 - 0.3 0.3 -
PVP K-30/90 (Povidone) 3.5 3.0 2.5 - - 2.2 2.2 -
Pregelatinized Starch NF - - - 15.0 - - 5.0
Hydroxypropyl Cellulose NF
Na lauryl sulfate/
Polysorbate 80
Sodium Bicarbonate USP
Aerosil
(Colloidal Silicon Dioxide NF)
-
-
-
0.3
-
-
-
-
-
-
-
0.5
% -
-
-
-
3.0
1.0
-
2.0
-
-
-
-
-
-
-
-
-
-
-
-
TOTAL PERCENTAGE Ü 100 100 100 100 100 100 100 100
Tablet weight (mg) Ü 380 300 400 660 650 275 550 380
Hardness SCU (target) 8-11 9 9 8-11 9 8-11 8-11 8-11
Disintegration <15 MIN 5-15 5-15 5-15 5-15 5-15 5-15 5-15 5-15
KEY:-
[I] Naproxen Na [J] Verapamil [K] Trazodone [L] Norfloxacin
[M] Norfloxen [N] Naproxen [O] Naproxen [P] Labetalol
ÜACTIVE MATERIAL %Ü 20.0 4.0 77.0 25.0 25.0 22.0 10.0 10.0
50.0
20.0
-
30.0
60.0
10.0
20.0
-
30.0
-
-
85.0
-
-
140
-
25.0
-
15.5
98.5
-
10.0
-
12.5
-
-
-
-
141
-
-
20.0
-
34.2
-
20.0
-
-
PVP K-30 (Povidone USP) 4.2 4.0 - 12.0 7.5 - 4.5
Hydroxypropyl Cellulose NF
- - 3.6 - 5.0 1.5 - -
Hydroxypropyl Methylcellulose
ALCOHOL 95% 32.0 32.0 85.0 28.0 18.0 25 10.0 20.0
Aerosil 1.0 1.0 5.0 1.0 0.5 0.5 0.5 0.5
(Colloidal Silicon Dioxide NF)
Talc 3.2
Sodium lauryl sulphate NF - - - - - - - -
Antioxidant - - - - - 0.1 - -
Solubilizing Agent - - - - - 0.2 - -
Citric acid : Ascorbic (2:1) - - - - - - 7.5 -
Organic Acids (15:1) 6.5
Magnesium Stearate 2.8 2.5 - 1.5 1.0 0.8 1.0 0.8
TOTAL PERCENTAGE 100 100 100 100 100 100 100 100
Tablet weight (mg) 250 250 970 300 200 480 200 200
Hardness SCU (target) 8 8 9 7-9 7-9 8-11 8-11 8-11
Disintegration <15 MIN 5-15 5-15 5-15 5-15 5-15 5-15 5-15 5-15
KEY:-
[A] Diclofenac K Tab [B] Omeprazole Tab [C] Nabumetone Tab [D] Diclofenac Na Tab
[E]Diclofenac Na Tab [F] Bupropion [G] Simvastitin Tab [H] ISMN 20mg Tab
References: Handbook of Generic Development Tablets IR
Vol. 1 Edition II (1999), Locum Press
Purified
problem becomes more acute.
The optimum scenario is for the R&D
department or development unit to be
connected, where possible to the same
Water water supply system as the commercial
production unit or alternatively install a
An Ingredient of Solid Dosage purified water system that mimics the
Form Development. proposed production quality, scheduled
for use in the full-size commercial lots.
‘develop all experimental batches This implies that the experimental
with batches, the scale-up lots, the process
Purified Water USP or Ph. Eur. -
similar or identical to the proposed qualification, pivotal and the initial three
commercial production quality.’ validation batches will all be prepared
and manufactured with commercial
production quality water.
T he use of appropriate water quality
as a non-active ingredient in the
development of pharmaceutical
The cleaning validation protocol
developed during the R&D phase
dosage forms, especially solid oral should use the same water quality.
granulating and coating solutions is
often realized at a late development
stage - occasional too late to be
Changing the
included in the product’s overall water quality during
development. product development
This section deals with the pitfalls of
failing to establishing strict criteria for may compromise
water quality both during pre- earlier results
formulation, formula development and
designing the cleaning validation Water quality for product development
protocol. should be at least purified water of
pharmacopoeial grade (USP / Ph. Eur)
Take a second look at and used from the very onset of the
development studies.
water as a key
The research or development water
non-active ingredient specifications should not change as the
for solid dosage form product develops and as the awareness
grows, that microbial parameters and
development pH are significant formula parameters,
to be evaluated in the development
Equivalent Commercial Quality process. Preparation of aqueous film
The development unit’s water quality coating solution during scale-up and
should be equivalent to the water early stability trials requires commercial
quality that is available for the quality purified water NF/BP
manufacture of the proposed Water-soluble semi-actives used (i.e.
commercial product. Where preservatives, anti-oxidants, chelating
development is separated from agents and coating dispersions) require
manufacture (in an overseas facility) formulation in a stable aqueous phase
the with standard and consistent
specifications.
ØCHECK LIST ×
SOP # P-HGD-01-Y2K
2. The latest Purified Water USP changes in the last Pharmacopeial qYesqNo
supplement has been checked for monograph changes?
3. The Purified Water used during the product development process qYesqNo
meets the requirements of the USP or Pharm Eur. monograph?
4. Each water test result falls within the USP/Pharm Eur. limits as qYesqNo
stated in the official monograph ?
ØCHECK LIST ×
SOP # P-HGD-01-Y2K
‘…review water quality batch lots for microbial levels - in real time…’
11. Careful note is taken of the microbial alert and warning levels in force at the qYesqNo
proposed commercial site?
12. The firms water quality alert / action levels are set at NMT 30 qYesqNo
organisms/mL?
13. The firms water quality warning levels are set at NMT 70 organisms/mL? qYesqNo
14. The firms water quality used for granulating solutions or coating suspensions qYesqNo
are set at maximum level of NMT 100 organism / mL?
15. The presence of frequent found organisms at the proposed manufacturing qYesqNo
site are noted and incorporated during product development testing?
16. The indicator microbe pseudomonas statzeri is used during microbial qYesqNo
limit test Studies during the solid oral dosage form development stages?
17. Months of consecutive commercial quality water lots have been evaluated qYesqNo
for upper and lower bioburden parameters ?
18. The final end-formula when spiked with water quality at the alert or qYesqNo
maximum levels adequately meets the microbial limit test (if required)?
19. Bulk ingredient water is boiled (30 min) and cooled to the required qYesqNo
temperature immediately before use, when used in open type granulating kettles
for granulation fluid preparations?
20. Bulk water when not in immediate use, is maintained at around 80o C and qYesqNo
continually circulated in appropriate distribution piping (closed system)?
21. The company maintains a full batch-to-batch analysis profile (in real time) of qYesqNo
all water quality lots produced for product production use?
22. All alert/action and warning level results from the batch-to-batch analysis are qYesqNo
fully investigated and proper corrective action taken?
Active Ingredients
‘Now think about the active ingredient, its source, its supply
and how to keep its specifications constant.’
Active Ingredients
Remember, BPC companies are DMF or GMP deficiencies cited by the
inspected by the FDA and the bulk FDA?)
chemical manufacturer’ active drug Don’t use a firms active drug
substance your generic firm may substance it you are not given full and
require or the bulk firms GMP may be detailed specifications and satisfactory
cited for a series of regulatory or drug replies to your DMF and GMP queries
master file (DMF) deficiencies. (Request the firms last two EIRs from
These deficiencies are your concern, the FOI services.)
should you choose to use their active Don’t be the first to reference the
drug substance for generic active drug in your ANDA unless a
development. Any DMF deficiencies thorough investigation has been made
at the time of ANDA submission will and all the active drug specifications
affect and delay the approval process are satisfactory.
of your generic ANDA file.
Don’t forget to place two or more
It is essential for the generic developer active drug batches (different lots #’s)
to communicate with the BPC firms’ on stability (40oC/3 months) in order to
regulatory affairs department and obtain a complete stressed analytical
request a summary of outstanding profile (examine all extraneous HPLC
DMF and GMP deficiencies specific to peaks obtained and cross check
the particular active drug required for unidentified peak ID with the active
your generic drug product manufacturer).
development. Part two of the
Handbook contains model systems of Don’t ever mix two lot/batch numbers
all the vendor / developer of the active drug substance for a
documentation requirements. developmental, analytical, pivotal or
validation study, due to insufficient
Do’s and Don’ts:- material.
Do insure that the batch lots of active Don’t use active material older than
drug are commercial batches.
18 months to two years for analytical,
Do obtain the drug’s active analytical stability or impurity studies, as non-
profile from the manufacture (for representative and non-comparative
commercial batches). data will be obtained.
Do request and obtain the drug active Don’t forget to obtain a statement
physical specifications on bulk density
from the DMF holder (the active drug
and particle size, (obtain analytical
manufacturer) that all issues
methods used to determine these
communicated to the DMF holder by
parameters).
the FDA have been fully addressed i.e.
Do get a analytical batch analysis of all deficiencies in the active DMF have
the last 6-10 consecutive commercial been corrected and at the time of
batches. writing there are no outstanding
Do inquire about the regulatory status deficiencies. These corrections are
of the firms DMF (has the DMF been also specific to the samples of
referred to previously by other ANDA commercial batches your firm has
applicants and are there outstanding received for development studies.
ØC H E C K L I S T ×
CL # P-HGD-02-Y2K
2. Each analytical test falls within the USP/NF limits as stated in the official qYes qNo.
USP monograph?
3. If the USP active material monograph is in the BP as well then the active qYes qNo.
passes the BP ‘Related Substances’ monograph test?
4. IR and UV Identification spectra (as per the USP/NF Identification test) qYes qNo.
has been supplied by the manufacturer?
6. All spectra are fully labeled and show the active materials lot # ? qYes qNo.
7. A letter of authorization obtained from the active supplier (DMF holder) qYes qNo.
addressed to the ANDA applicant referencing the active DMF #, as used in
the ANDA generic product?
8. The DMF holder (active manufacturer) has included a statement that all qYes qNo.
FDA issues / concerns communicated to the DMF holder i.e. deficiencies in
the active DMF have been fully addressed?
9. The active material safety data sheet has been provided by the approved qYes qNo.
supplier?
10. The latest USP supplement has been checked for monograph changes? qYes qNo.
Footnote : Checklist applies to non compendial (non USP) active drug substances - where applicable .
Active Ingredients
‘…timeline for active materials from first look to Approved Supplier…’
ØC H E C K L I S T ×
CL # P-HGD-02-Y2K
9. Vendor DMF and GMP issues have been fully resolved qYes qNo.
11. A full batch-to-batch analysis with three lot #’s has been tested? qYes qNo.
12. All abnormal results from the batch-to-batch analysis qYes qNo.
investigated
13. Are the accelerated stability results of the active drug qYes qNo.
substance alone and in the proposed formulation approvable ?
14. Does the purchasing department have a full set of approved qYes qNo.
specifications ?
15. Has a vendor approval certificate for the approved active been qYes qNo.
issued (copy with purchasing department)
STANDARD OPERATING
PROCEDURES
SOP # P-HGD-02-Y2K
ACTIVE INGREDIENTS
P-000-02- Y2K Active Drug Substances for Generic Drugs.
P-000-02- Y2K Active Drug Substances specifications procedure prior to purchasing.
P-000-02-Y2K Vendor Certification Requirements for Approved Actives.
P-000-02-Y2K Decision tree for establishing impurity acceptance criteria.
P-000-02-Y2K Decision tree for establishing degradation acceptance criteria.
P-000-02-Y2K Decision tree for establishing particle size acceptance criteria.
.P-000-02-Y2K Decision tree for establishing polymorphism existence.
.P-000-02-Y2K Decision tree for establishing microbiological testing.
.P-000-02-Y2K Decision tree for evaluating chiral actives.
P-000-02-Y2K Developing Product Formula with Approved Actives.
P-000-02-Y2K Inventory Records for the Active Drug Substance.
P-000-02-Y2K Investigating and handling abnormal batch results.
1
Solid State Forms Some drug substances exist in different solid state forms
(polymorphs or solvates) which differ in their physical properties. Differences in
these forms have been shown to affect drug product performance, dissolution
bioavailability and stability.
1
Polymorphism: the occurrences of different crystalline forms of the same drug
substance. This may include solvation or hydration products (also known as
pseudopolymorphism) and amorphous forms.
Physico-chemical measurements and techniques are commonly used to determine
whether multiple forms exist. Examples of these procedures are:
n Melting point (including hot stage microscopy)
n Solid State Infra Red Common tests
n X-Ray Powder Diffraction in Generic
n Thermal Analysis (DSC ; TGA ; DTA) Actives
-------------------------------------------------------------
n Raman Spectroscopy
n Scanning Electron Microscopy Less
n Solid State NMR common
procedures
4
[End of Document]
'Semi Actives'
‘…Challenge each preservative and anti-oxidant system during
development and then optimize its formula concentration…’
ØC H E C K L I S T ×
CL # P-HGD-03-Y2K
3. Does the of the antioxidant percent chosen represent the lowest % value q Yes qNo
to minimize impurities and degradants during the inferred product shelf life?
4. Has the overall excipient formula been evaluated for total heavy metal q Yes qNo
content from the inactive C of As and product specification data sheets?
5. Have reducing agents, antioxidant synergist and sequestering agents that q Yes qNo
have not been appropriately validated, been excluded from the product
formula?
6. Does the active ingredient remain in the check specification range (90.0 - q Yes qNo
110.0 of labeled amount) at the end of accelerated stability testing?
7. Does the potency of the active ingredient decrease when the chelating q Yes qNo
agent is removed from the product formulation during normal and aging
studies?
8. Have range studies been performed to evaluate the optimum amount? qYes qNo
9. Has the product formula been evaluated at lower, middle and upper q Yes qNo
antioxidant percentages and evaluated against active assay values?
10. Has it been clearly established that the inclusion of chelating or an q Yes qNo
antioxidant synergist positively enhances the action of the antioxidant?
11. Has potency loss of the semi actives been fully demonstrated during the q Yes qNo
product development stages to establish valid specification ranges?
12. Does the stability testing protocol only evaluate formula specifications q Yes qNo
that are directly impacted by the aging process ?
13. Has a complete product development profile of the antioxidant been q Yes qNo
evaluated in order to eliminate routine release and stability testing of the
antioxidant agent during commercial manufacture ?
14. Is the stability testing protocol for the pivotal batch a logical q Yes qNo
development sequence from the product development work ?
Footnote :
Bold numbers in checklist indicate this work must be qualified and or validated before
manufacturing the Process Qualification Batch, which actually marks the end of the product
development stage.
Qualifying
Observations and Results:
⇒ Batch P-04 manufactured without
Cremophor displayed, higher hardness, a
short disintegration time and good
the ANTIOXIDANT stability results compared with batches
manufactured with Cremophor™.
T he qualification of the antioxidant
concentration in the formula is shown
here as an example in order to
⇒ Observations during the tabletting
process showed that batches formulated
with Cremophor™ did not achieve the
evaluate the effect of increasing target hardness, thus the use of solubility
quantities of the antioxidant Butylated agent for BHA is thereby eliminated
Hydroxyanisole.
BHA ranging from zero (acting as a Stability tests demonstrated that:
control) to a maximum of 0.15 percent of ♦ Control Batch, P-05 manufactured
the tablet weight is incorporated into a without BHA showed an significant
250 mg immediate release oral tablet increase in the percent of impurities /
formulation (Ref. Table No. 01). degradation products after the one month
interval. (Refer Table 7 - shaded area)
The use of Cremophor™ RH40 (ranging
♦ Formulations containing BHA
from zero percent to a maximum of 0.5%
antioxidant quantities between 0.05%
of the tablet weight) is added to evaluate
(0.195mg) and 0.15% (0.580mg) of the
a possible improvement in the Butylated
tablet core weight, afforded adequate
Hydroxyanisole NF solubility during the
stability results for Assay and Impurities.
manufacturing stages.
♦ As thin layer chromatography and
The granulation is performed via a High supporting HPLC impurity analysis
Shear Granulator (Diosna P-10™) using demonstrates that similar results were
dry PVP K-30™ as the tablet binder and obtained, it is concluded that presence of
Purified Water USP as the granulation BHA serves as an antioxidant that inhibits
agent. Microcrystalline Cellulose is and retards the growth of impurities and
incorporated extra-granularly. degradants.
The tablet cores are coated in the Accela Based on the above antioxidant
Cota™ A10 and the film-coated tablets, qualification experiments the choice of
packaged in High Density Polyethylene formula, P-04 (0.1% BHA) was selected
containers, which are evaluated for for further in the optimization and
stability at accelerated stability conditions qualification batch development:
of 40°C / 75% RH for 3 months. Stability Chosen Formula:
Indicating tests methods (TLC and HPLC)
used for investigating Assay, Dissolution • Active material 250 mg
and known Impurities are fully validated.
• PVP K-30 - (Dry binder)
The physical parameters of the • Microcrystalline Cellulose NF - (Dry Filler)
granulates tested are shown in Table No.
02 and the granulates for all batches • Dry Starch NF (Disintegrant)
demonstrated adequate flow and • BHA (Antioxidant 0.1%)
compressibility characteristics. The • Purified Water USP (Granulation fluid)
physical test results for the tablet cores
are presented in Table No. 03. • Stearic Acid NF (~1.4 - 1.5%)
The optimization batch stability results • Magnesium Stearate NF (~0.25%)
are presented in Table No. 04 - 07
.
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
Optimization Batch Stage
Table No. 01.
Formulae Ingredients Amount per tablet core (mg)
ò
P-01 P-02 P-03 P-04 P-05
Batch No. è
Part I - Pre-mix (dry) High shear mixing Control Control
Active Material 250.0 250.0 250.0 250.0 250.0
Microcrystalline Cellulose NF 30.0 30.0 30.0 30.0 30.0
(Avicel PH101™)
Povidone USP 8.0 8.0 8.0 8.0 8.0
PVP K-30™
Starch NF 40.0 40.0 40.0 40.0 40.0
Part II - Granulation solution Granulating Stage
Butylated Hydroxyanisole NF 0.195 0.580 0.390 0.390 -
(BHA). (0.05%) (0.15%) (0.1%) (0.1%) (Control)
Polyoxyl 40 Hydrogenerated 2.0 2.0 2.0 - 2.0
Caster Oil NF. (Control)
(Cremophor RH40™)
Alcohol USP (95%) Qs Qs Qs Qs Qs
Purified Water USP Qs Qs Qs Qs Qs
PART III - Extra-granule Dry Mixing
Microcrystalline Cellulose NF 52.8 52.4 52.6 50.6 53.0
(Avicel PH102™)
PART IV - Lubrication Rapid Dry Blending
Stearic Acid NF 6.0 6.0 6.0 6.0 6.0
Magnesium Stearate NF 1.0 1.0 1.0 1.0 1.0
Total Core Weight 390.0 390.0 390.0 390.0 390.0
Qualification of Antioxidant
Table No. 02.
Hardness 8 9 8 13 9
Average of 10 (SCU)
Disintegration Time 1:55 1:47 1:50 1:31 1:33
Average of 6 (min)
Friability (%) 0.21 0.16 0.2 0.15 0.15
COATED TABLET
Disintegration Time 2:55 2:50 2:35 2:26 3:05
Average of 6 (min)
P-04 &- P-05 Control Batches.
Reference:
Carr’s Index - Compressibility = (CVD-BD) / CVD×100) Wells, J.I. - Pharmaceutical Preformulation -
Chapter 7: Powder Flow Properties.)
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
Specs. White to Off NMT 90.0 - NMT NMT NMT NMT NMT NMT NMT NMT
White 15 110.0 1.0 0.5 0.5 0.1 0.5 0.1
3.0 1.0
0 Conforms 2’54” 102.0 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 <0.02 <0.04
1 Conforms 3’09” 99.8 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 0.02 0.04
2 Conforms 2’46” 101.2 <0.1 <0.1 0.1 0.1 <0.3 0.05 0.05 0.10
3 Conforms 2’43” 100.8 <0.1 0.1 0.1 0.1 0.3 0.1 0.05 0.15
Key:
Assay % of Label Claim of Active BHA - Butylated Hydroxyanisole NF Dissolution % of Label Claim of Active
ANY I = Any other impurities (by TLC) TOTAL I = Total impurities (by TLC) ANY II = Any other impurities (HPLC)
TOTAL II = Total impurities (by HPLC)
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
Batch No: P01
Specs. White to Off NMT 15 90.0- NMT NMT 0.5 NMT 0.5 NMT 0.1 NMT 3.0 NMT 0.5 NMT 0.1 NMT 1.0 NLT 75% (Q)
White 110.0 1.0 dissolved in 30’
0 Conforms 2'.45'' 102.8 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 <0.02 <0.04 92.6 98.1 96.9
(5.1) (3.0) (4.5)
1 Conforms 2'..34'' 100.9 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 <0.02 <0.04 98.0
(2.1)
2 Conforms 2'..45'' 100.9 <0.3 <0.1 <0.1 <0.1 <0.5 <0.02 <0.05 0.07 99.8
(2.2)
3 Conforms 2'':55'' 102.1 0.3 0.1 0.1 0.1 0.5 0.06 0.05 0.11 95.1 97.2 96.7
(3.2) (2.4) (1.4)
Table No. 05
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
0.15% Antioxidant - Test Batch
Batch No: P02
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
No Antioxidant - Control Batch
Batch No: P05
Specs. White to NMT 15 90.0- NMT 1.0 NMT 0.5 NMT 0.5 NMT 0.1 NMT 3.0 NMT 0.5 NMT 0.1 NMT 1.0
Off White 110.0
0 Conforms 2’40” 100.8 <0.1 <0.1 <0.1 <0.1 0.1 <0.03 <0.02 <0.05
1 Conforms 2’50” 99.8 0.2 0.1 0.1 0.1 0.4 <0.03 <0.62 <0.65
2 Conforms 2.55” 101.8 0.6 <0.1 0.2 0.2 1.1 <0.02 <0.58 0.60
3 Conforms 2.40” 102.2 1.4 <0.2 0.2 0.21 2.0 <0.04 <0.84 <0.88
Table No. 07
Note: Control Batch - No dissolution studies necessary High Impurity Significant
value indicate Increase in
antioxidant impurities
requirements
Qualification of Antioxidant
Oral Tablets 250 mg Active Material
Specs. White to NMT 15 90.0- NMT 1.0 NMT 0.5 NMT 0.5 NMT 0.1 NMT 3.0 NMT 0.5 NMT 0.1 NMT 1.0
Off White 110.0
0 Conforms 2’55” 101.8 <0.1 <0.1 <0.2 <0.1 0.5 <0.03 <0.05 <0.08
1 Conforms 2’40” 100.7 0.2 0.1 0.1 0.1 05 <0.03 <0.05 <0.08
2 Conforms 2.50” 100.8 0.5 <0.2 0.2 0.05 0.95 <0.02 <0.05 0.07
3 Conforms 2.50” 103.9 0.6 <0.4 0.3 0.1 1.4 <0.04 <0.10 <0.14
Table No. 07B
Note: Reference Batch - Dissolution studies necessary
Impurities
serve as a
REFERENCE
ØC H E C K L I S T ×
CL # P-HPGD-03-Y2K
1. Has the RLD’s non active ingredients been qualitatively identified ? qYes qNo
2. Is the generic formulation a basic Q&Q formula of the RLD ? qYes qNo
3. Are the non actives referenced in the FDA’s IIG Guide qYes qNo
4. Has the maximum percentage not been exceeded for oral tablets? qYes qNo
5. Has the particle size and bulk density of key non actives (e.g. qYes qNo
lubricants) been specified with an appropriate range?
6. Is the dissolution profile of the proposed generic formula similar to qYes qNo
the RLD's profile?
7. Are the comparative 12 point dissolution values all within 5% of the qYes qNo
RLD dissolution profile under normal and accelerated testing?
8. Is the granulation uniformity of content spread less than 4.0 - 5.0% qYes qNo
with RSD ,6.0%?
9. Does the development protocol indicate that the final formula is qYes qNo
manufactures at the lower and upper tabletting speeds ?
10. Does the firm regularly review the Pharmacopoeial Forum for qYes qNo
proposed monographs and specifications for non-compendial
excipients ?
11. Has the firm reviewed all the suppliers for potential ‘Approved qYes qNo
Suppliers’ as listed the Handbook of Pharmaceutical Excipients ?
12. Is Purified Water USP used as an approved excipient granulating qYes qNo
agent and coating suspension ingredient?
13. Have the excipient specifications been reviewed in USP / NF, qYes qNo
Ph. Eur / BP, and JP and the latest supplements and addenda?
14. For compendial excipients has the latest supplement been qYes qNo
checked ?
15. Does your generic firm have a current ‘ Approved Supplier SOP ' qYes qNo
for non active ingredients?
Footnote : The words non active ingredient; inactive ingredient and excipient are all the same meaning and interchangeable in use.
1
GRAS - Generally Accepted As Safe (Ref. 21 Code FR )
STANDARD OPERATING
PROCEDURES
CL # P-HPGD-03-Y2K
4
[End of Document]
Container-Closure Systems
‘…the container documentation is the key
dot every i and cross every t…’
ØC H E C K L I S T ×
CL # P-HGD-03-Y2K
Container-liner-closure Systems
‘…check that the suppliers specifications - always refer to the latest USP edition…’
1. The components of the thermoplastic tube complies with the 21 qYes qNo
CFR?
3. HDPE complies with the USP requirements for HDPE ? qYes qNo
6. The inner seals in the cap complies with the 21 CFR 176.180 qYes qNo
7. The vendor’s name & address of each component is identified ? qYes qNo
8. The manufacturer of the tube resin cap and liner etc. is identified? qYes qNo
9. The resin type and code number is fully identified ? qYes qNo
10. Specifications comply with the current USP requirements ? qYes qNo
11. Each pack size (e.g. 20, 50, 100 units) has a separate set of qYes qNo
component specifications (for container, resin, liner, cap, colorant etc.)
12. Letters of authorization (LOA) from the DMF holders obtained? qYes qNo
13. Certificates of Compliance identifying the materials used in the qYes qNo
component manufacture conform to 21 CFR 172-177 etc.?
14. Certificate of Analysis identifying component meets specific USP qYes qNo
Test Requirements (e.g. moisture permeability test etc.)?
15. GMP Certification statement indicating GMP status of component qYes qNo
manufacturing facility?
Footnote : Bold numbers in checklist indicate that this documentation must be on file with generic drug
developer prior to process qualification or pivotal batch manufacture.
ØC H E C K L I S T ×
CL # P-HGD-03-Y2K
Container-Liner-Closure Systems
♦ All materials are manufactured in accordance with DMF # 1234 qYes qNo
♦ No materials specification changes will be made without full notification to all qYes qNo
ANDA holders ?
17. Technical data sheet and component drawings supplied by manufacturer qYes qNo
for each component (container, liner composite, closure, fillers)?
18. Separate component drawings and specifications for - container? qYes qNo
19. Separate component drawings and specifications for - liner/laminate? qYes qNo
20. Separate component drawings and specifications for - cap / closure? qYes qNo
21. Separate specifications and certificate of analysis for fillers (cotton wool) qYes qNo
22. Generic Developer has a LOA for every DMF File referenced to FDA? qYes qNo
23. All DMF #’s referenced in ANDA are identified on ANDA cover page? qYes qNo
24. Documentation has been sourced on time during product design phase ? qYes qNo
25. Documentation has been audited in-house for current pharmacopoeial qYes qNo
editions, latest Cert. of Analysis and updated component specifications ?
26. Packaging system complies with 'Guidance for Industry Application for qYes qNo
Container Closure Systems used for Packaging of Human Drugs' (Draft 6/1998)
Footnote : Insure that vendors and manufacturers have updated all their component specification and
documentation to the current year. Specifications must apply to actual batch lot supplied to developers.
STANDARD OPERATING
PROCEDURES
CL # P-HGD-03-Y2K Page 1 of 1.
Container-Liner-Closure Systems
The Attached SOP for a ANDA drug products highlights the components quality
control specifications and documentation requirements for filing an drug product
application. The following selected model Standard Operating Procedures are
recommended SOPs :
CONTAINER-LINER-CLOSURE SYSTEMS
# P-000-01-129Y Container-liner-closure systems for generic development.
# P-000-01-129Y Documentation requirements for Container-liner-closure systems .
21 CFR references:-
The Indirect Food Additive regulations in the 21 Code Federal Register (21 CFR)
that are applicable to solid oral dose container closure packaging components are:-
21 Code Federal Register (21 CFR 170 -199) - Food Additive Regulations
Indirect Food Additives General - Part 174
Indirect Food Additives Adhesives and Coatings - Part 175
Indirect Food Additives Paper & Cardboard - Part 176
Indirect Food Additives Polymers - Part 177
Indirect Food Additives Adjuvants / Sanitizers - Part 178
Indirect Food Additives GRAS - Part 186
Examples;
Colorant (in thermoplastic) - 175.300
Resinous & polymeric coatings - 175.300
Adhesives - 175.105
Paper and paperboard (contact aqueous/fatty) - 176.170
Inner seals (in closures/caps) - 176.180
Fluorocarbon resins - 177.1380
Olefin polymers - 177.1580
Polyethylene phthalate polymers - 177.1630
HDPP (Polypropylene)1 - 177. 1520
HDPE (Polyethylene) 2 - 177.1520
HDPE Resins (of thermoplastic) - 177.1520
- 177.280 (175.300)
1 High Density Polypropylene - HDPP
2 High Density Polyethylene - HDPE
4
[End of Document]
Manufacturing
Instructions
'…start with the order of addition and the processing conditions...
end with process optimization and process qualification...'
Altering the dissolution parameters: the lubricant volume and aid in the
Without formula changes dissolution uniform coating of the granules during
may be altered by several factors such blending.
as (a) the order of addition or (b) Over lubrication and non-uniform
varying the amount of intra-granular lubrication of the granules may not be
and extra granular disintegrant as well detected in the tabletting process but
as (c) controlled lubrication coating via will manifest itself in the consistency of
short or longer blending times of the the batch-to-batch dissolution analysis
finished granule with the lubricating which may impact on the
agent / glidant combination. The bioequivalency testing.
addition of Dry Starch NF at the Compress the final development
lubrication stage (in the blender) formula or the process qualification
improves the tablet dissolution by batch on the actual commercial
partial coating of the lubricant(s). machine at the highest tabletting
Granulating Agent - Dry or Solution speed to insure the absence of
The Addition of the binder (granulating compression problems (capping,
agent) e.g. PVP K30™ may be added screeching etc.)
as an initial dry mix with the active or
alternatively dissolved in solution with Factors impacting on dissolution
the granulating fluid (e.g. water or profile:
alcohol) an added (rate) to the ◊ Active particle size and bulk
granulating vessel.
density
Granule moisture range
The amount of moisture in the granule ◊ Excipient formulation of non-
when it exceeds the range 2.0 - 2.5% actives
results in harder tablets on aging with ◊ Order and stage of addition
longer dissolution times. Control of ◊ Intra-granular disintegrating agent
granule moisture and comparison of quantity and blending time (Y-
resulting tablet dissolution is a critical Cone).
development parameter. Improved ◊ Inter-granular disintegrating agent
dissolution can be obtained by quantity and blending time (Y-
reducing moisture to optimum levels cone).
that result in elegant cores or tablets.
◊ Granulation moisture content.
Granule blending times ◊ Quantity of lubricant in formula.
Control of the granule ‘coating’ ◊ Splitting blending amounts and
process of the lubricant (i.e. Mg blending times - for lubricants.
Stearate and/or Stearic Acid) may be
◊ Lubrication blending times - 3-5
achieved by a two stage lubricating
blend. When the lubricant blending
minutes generally (Y-cone)
times are reduced, the lubricant should The formula given in Chapter Five and
be bulked up (with a reserve a portion Charts 1 to 3 use most of the above
of cornstarch) to eliminate partial over- development parameters to produce a
coating of the granules in initial drug product comparative to the RLD
contact with resulting non-uniform that shows excellent stability and long
coating of the entire granulation term stable impurity profiles.
material. A premix of the glidant and
lubricant can be considered. Flow charts for Dry and wet
Lubricants may also be blended with granulation are given to highlight the
extragranular disintegrant to bulk-up various manufacturing techniques
Process Validation
Developing the manufacturing process ♦ mixing speeds (rpm; chopper I / II)
and process parameters will result in a ♦ mixing times (start / stop)
rugged set of manufacturing
instructions and in-process controls. ♦ FBD parameters (drying times)
The end point of the process ♦ Granule LOD content.
optimization is a consistent batch-to- ♦ Addition of disintegrants to FDB or
batch product quality and uniform milling stage
finished product specifications.
♦ blending parameters and order of
In order to achieve consistent batch- addition of lubricants and glidants
to-batch analysis, the manufacturing
♦ Multiple lubricants (Mg Stearate
procedure requires individual process
0.25% and Stearic Acid 0.75%)
step optimization (i.e. process
validation). ♦ Short lubricant blending times
Manufacturing Instructions ♦ Partial coating of lubricants with
glidants and/or Disintegrants
Optimize the manufacturing process.
The process optimization procedure ♦ maximum processing times per step
requires individual process step Development and optimization
evaluation during the development of
From this development and
the process manufacturing
optimization procedures - key process
instructions.
steps can be identified with the
The end of the product development potential to affect the finished drug
phase will result in a regulatory product both in quality and
demonstration of the development specifications. It is these key
optimization procedure or development processing steps that are addressed
validation with the demonstration of and demonstrated in the validation
the pivotal generic batch and protocol for the initial three,
thereafter the three consecutive consecutive, full size, validation
validation batches for commercial batches.
sale. Upper and lower in-process controls
The manufacturing process requires are established for these critical
certain key steps to be evaluated for processing steps and these limits are
optimizing the process and producing individually challenged and qualified
a rugged end product. under the product development
These development steps are: program.
Addition of ingredients- The final regulatory demonstration of
all operating ranges and product
♦ Screening of bulk powders (size)
specification limits takes place with the
♦ Pre-blending procedures for small three commercial validation batches.
amounts (plastic bags prohibited) Only parameters that can impact on
♦ Order of addition of excipients the product quality are evaluated in a
validation program.
♦ Dry mixing the binder in Granulator This process of developmental
♦ Addition rates times for granulating validation and exhibition validation
fluids. results in a final validated process
Processing of the ingredient mix producing a rugged drug product
meeting the desired drug product
♦ High/low shear mixing settings
specifications.
ØC H E C K L I S T ×
CL # P-HGD-03-Y2K
Manufacturing Instructions
(example of a wet granulation procedure)
1. The manufacturing instructions specify the exact equipment used ? qYes qNo
2. Major equipment is identified with an ID number ? qYes qNo
3. The words “use suitable / appropriate... etc. ” are not used ? qYes qNo
4. Mixer/granulator times have a ‘start and stop’ recording time ? qYes qNo
5. Mixer/granulator speeds and chopper settings are precisely stated ? qYes qNo
6. Instructions identify the mixing times and temperatures for preparing the qYes qNo
granulating solution?
7. Operator and check signatures are per individual step ? qYes qNo
9. Manufacturing steps are broken down to one action only ? qYes qNo
10. FBD temperatures are recorded as T0 C ( ± 2/3 0 C) - avoid ranges ? qYes qNo
11. FBD inlet, outlet and bed temperatures are recorded ? qYes qNo
12. Drying is continued until the target granulate moisture percentage qYes qNo
determined by LOD (± 0.2 - 0.4%) is reached ? Record overall drying time.
13. Dried granules are milled stating screen size and machine settings ? qYes qNo
14. Blender rpm and blending time recorded (with / without bar)? qYes qNo
17. The maximum standing time before compression is indicated ? qYes qNo
18. Machine speed is stated (target, lower and upper rpm) ?. qYes qNo
19. The sample size, number of samples and sampling location (i.e. LHS and qYes qNo
RHS) is identified in batch sheets?
Footnote : Where possible transfer the lubricated granules into a double polyethylene lined drum. Close well and protect from light where
necessary using an inner transparent bag and an outer black bag. Obtain the granule net weight and calculate the yield.
Manufacturing
Instructions
Manufacturing and Controls
BATCH MANUFACTURING INSTRUCTIONS
A DETAILED EXAMPLE
NOTE:
The concept of a single batch
Edition Number: Effective Date: APPROVED
01
means;
Ed. Status MM/DD/199Y _____________ __________Granulation - Blend -_________/________
_______________ Coating
New Department R &D 1 - 3 RA : ONE : 1 - QC 4 / QA
[INGREDIENT #1 NF]
[INGREDIENT #2NF]
[INGREDIENT #3 NF] NOTE:
[INGREDIENT #4 NF] The ingredients are written in
MIX for [ 0 ] minutes at mixer speed [I] or [II] and Chopper [I] or [II] the same order as they are
JB and
processed LJ also as they
appear in the 'Master
Formula'
3. Granulation Preparation - No. I
5. OPERATE the mixer and add the [GRANULATING AGENT USP] and
mix until fully dissolved. JB LJ
6. LOAD the [High Shear Mixer 000] (Type & No) with the following JB LJ
ingredient :
7. MIX for [0] minutes at mixer speed [I / II] and Chopper [I / II]. JB LJ
11. DRY the wet granulate in the FBD (Type & No) under the following JB LJ
settings:
Time of Drying [00] min. Ñ 10%
Inlet Air Temperature NMT [00]º C (Target: [00]º C)
Outlet Air Temperature NMT [00]º C (Target: [00]º C)
12. ATTACH the temperature graph of the FBD (Type & No) to the JB LJ
manufacturing instructions.
Immediately ADD the batch number to the temperature graph and date JB LJ
and sign it.
13. MILL about 1Kg. 'check portion' the dried granulate through an JB LJ
OSCILLATING GRANULATOR (Type & No) fitted with a [0.0 mm] screen.
14. CHECK the milled granulate portion for Loss on Drying (LOD). JB LJ
Use (Type & No) IR machine with temperature set at temperature [00]º C
Record First result: __________[ 0.0%]
LOD Limits: [0.0 to 0.0%]
15. If necessary, CONTINUE to DRY the bulk granulate under the same JB LJ
conditions as stage 11, until the LOD is close to the midpoint of the given
range limits and check moisture again.
JB LJ
16. RECORD Second result: __________[ 0.0%]
17. PASS the Dried Granulate through the OSCILLATING JB LJ
GRANULATOR (Type & No) fitted with a [0.0 mm] screen into a [000]
liter container or bin.
JB LJ
18. WEIGH the milled granulate. ______Kg.
Immediately ADD the batch number to the scale print-out, attach to the JB LJ
manufacturing instructions, date and sign the print-out.
19. Theoretical Weight [00.0] Kg. Yield __________ %
(Yield Limits: NLT 95% of Theoretical Weight.) Bins __________
21. LOAD the [High Shear Mixer 000] (Type & No) with the following
JB LJ
ingredients in order of addition:
[ACTIVE INGREDIENT BP];
JB LJ
Mix for [ 0 ] minutes at mixer speed [I] or [II] and Chopper [I] or [II]
JB LJ
22. ADD the granulating solution to the [High Shear Mixer 000] (Type &
No) while mixing at mixer speed [II] and chopper speed [II].
Total Mixing Time is 45 seconds. JB LJ
Time of adding Solution/SPRAYING - [40] seconds
Time of mixing - [ 5] seconds
JB LJ
23. If necessary, ADD / SPRAY the [PURIFIED WATER USP]. q.s. and
mix at the same conditions as in stage 22.
Amount of additional [PURIFIED WATER USP]: __________ Kg. JB LJ
Additional mixing time __________ Seconds
24. DISCHARGE the wet granulate to the FBD mobile bed (Type & No) JB LJ
while mixing at mixer speed I.
25. DRY the wet granulate in the FBD (Type & No) under the following JB LJ
settings:
Time of Drying [00] min Ñ 10%
Inlet Air Temperature NMT [00]º C (Target: [00]º C)
JB LJ
Outlet Air Temperature NMT [00]º C (Target: [00]º C)
JB LJ
ATTACH the temperature graph of the FBD (Type & No) to the
manufacturing instructions. Immediately add the batch number to the
temperature graph and date and sign it.
JB LJ
26. MILL about a 1Kg. 'check portion' of dried granulate through a
OSCILLATING GRANULATOR (Type & No) fitted with a [0.0 mm] JB LJ
screen.
27. CHECK the milled granulate portion for Loss on Drying (LOD).
ED. N0: 02 Effective Date APPROVED:
Replaces Ed 01. :
Ed. Status: MM/DD/2000
Operational Department R&D QC QA
USE (Type & No) IR machine with temperature set at temperature [00]º C JB LJ
RECORD First LOD result ________ [0.0%]
LOD Limits: - [0.0 to 0.0%]
28. If necessary, CONTINUE to DRY the bulk granulate under the same
JB LJ
conditions as stage 25, until the LOD is close to the midpoint of the given
range limits and check moisture again.
39. SEAL the double PE plastic bags (clear inner, black outer) with JB LJ
plastic ties then close all containers, and attach (bar coded) labels to the
Bulk Containers for transport to the holding area.
PART FOUR
FILM COATING SUSPENSION
JB LJ
40. IDENTIFY and verify the cleanliness of the coating equipment.
Weigh [00] Kg PURIFIED WATER USP into a stainless steel vessel
fitted with a roller mixer. (Vessel # 0) JB LJ
41. ADD gradually, while mixing the [FILM-DRY OD-P-0000 - Color] to
the PURIFIED WATER USP and mix to a uniform dispersion - about 45
minutes:
Mixing time ________minutes.
42. PASS the AQUEOUS FILM COATING SUSPENSION through a [00] JB LJ
mesh screen into a stainless steel container and close well.
JB LJ
PREVENT
43. SPLIT the AQUEOUS FILM COATING SUSPENSION into equal
Foaming of
sublots and label with (bar-coded) batch number and Sublot number. aqueous dispersion
(Allow to de-aerate)
44. NB: STIR the AQUEOUS FILM COATING SUSPENSION
continuously during the coating process.
SPRAYING PARAMETERS
Pump type: Peristaltic
Spray rate: [000] - [000] g/min
Nozzles: [0] - [0.0] mm
INLET AIR
Angle of Guns to Bed: 90 degrees
Relative Humidity
Height above Bed: [00] cm
should be controlled
Incoming Air Temperature: [00]º C - [00]º C (Target: 00ºC) JBday-to-day
(to prevent LJ
Extraction Air Temperature: [00]º C - [00]º C (Target: 00ºC) environmental variation)
46. COAT tablet cores at a drum speed of [0] - [0] rpm and a spray rate JB LJ
of [000] - [000] g/min until the target COATED tablet weight of [000] mg
is obtained.
COATING COMPLETION PROCEDURE. JB LJ
47. REDUCE drum speed to minimum rpm and perform the following:
- Reduce set point of incoming air temperature to 00ºC
- on reaching this temperature - close the inlet air.
- continue drum speed until the Extraction Air Temperature reaches 00ºC
- 00ºC
48. TRANSFER coated tablets into containers lined with two PE plastic JB LJ
bags (clear inner, black outer). Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
49. ATTACH the temperature graphs (Type & No) to the manufacturing JB LJ
instructions. Immediately add the batch / Sublot number to the
temperature graph(s) and date and sign graph.
JB LJ
50. WEIGH Coated tablets
Actual production weight: [00.0] Kg. No of Containers
SPRAYING PARAMETERS
Pump type: Peristaltic
JB LJ
Spray rate: [000] - [000] g/min
Nozzles: [0] - [0.0] mm
Angle of Guns to Bed: 90 degrees
Height above Bed: [00] cm
Incoming Air Temperature: [00]º C - [00]º C (Target: 00ºC)
JB LJ
Extraction Air Temperature: [00]º C - [00]º C (Target: 00ºC)
52. COAT tablet cores at a drum speed of [0] - [0] rpm and a spray rate JB LJ
of [000] - [000] g/min until the target COATED tablet weight of [000] mg
is obtained.
COATING COMPLETION PROCEDURE
53. REDUCE drum speed to minimum rpm and perform the following: JB LJ
- Reduce set point of incoming air temperature to 00ºC
- on reaching this temperature - close the inlet air.
- continue drum speed until the Extraction Air Temperature reaches [00ºC JB LJ
- 00]º C.
JB LJ
54. TRANSFER coated tablets into containers lined with two PE plastic
bags (clear inner, black outer) Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
55. ATTACH the temperature graphs (Type & No) to the manufacturing JB LJ
instructions. Immediately add the batch / Sublot number to the
temperature graph(s) and date and sign graph.
M a n u f a c t u r i n g
[Matrix excipient USP]
Flow Chart I
[Matrix excipient NF]
[Matrix excipient NF]
BATCH MANUFACTURING FLOW CHART
[Generic name] Tablets [USP] [000.0] mg.
Step 1
Matrix Premix Blend [[Active Material] High Speed Granulator
Tumble mixer Matrix Premix Blend PREMIX
Step 2b
High Speed Granulator [Purified Water USP}
Extra-granular GRANULATION if required
Glidant Excipient
Step 4
Communition stage
Lubricant MILLING
Step 5
Quick Sieve Stage 5 Extragranular Addition
Sieve # [0] pre-blending
IPQC testing
Step 6 Blend Uniformity
Final Blend Twin Shell / Flow Bin Sieve Analysis
Yield Analysis Blending
Step 8
Coating stage IPQC testing
[1/2/3] Sublots coated tablets- weight
Coating Yields
Overall Prod Yield
Finished Product
Chart One:
M a n u f a c t u r i n g
Flow Chart II
Dry Granulation
Example of a Dry Granulation Manufacturing Process
Y-CONE 5, 10 MINS.
ACTIVE MATERIAL USP
STARCH NF (REDRIED)
TRITURATION SIEVE 40 MESH
CONTAINER
ANHYDROUS LACTOSE NF #1
CONTAINER #2
3
SIEVE 30 MESH
1 2
SIEVE 30 MESH
SIEVE 30 MESH
MAGNESIUM STEARATE NF
SIEVE 50 MESH
TABLETTING SLUGS
KILIAN PHARMA
SIEVE 50 MESH
Chart Two - Example of a Dry Granulation Manufacturing Process - Stages are Dry Mixing; Slugging;
Milling; Blending & Compression.
Manufacturing
Flow Chart II
GRANULATION
DIOSNA P-800
PVP K-90 (Povidone USP)
Mixing:
Purified Water USP
MIXER II +
CHOPPER II
S/S Mixing vessel 140 seconds
Propeller Mixer
DIOSNA P-850
Purified Water USP Mixing:
MIXER II +
CHOPPER II
DRYING - FBD
Outlet temp. up to 45°C MILLING
Dry until
(target 42°C) 0.8 mm screen
Target LOD Inlet temp. up to 68°C
Reached (target 64°C)
Y-CONE 200
Magnesium Stearate NF SCREENING 5 min. Mixing
50 mesh screen Discharging into drums
TABLETTING
Tabletting according to
specifications
RANITIDINE HCl
300mg
COATED CAPLETS
MF
AND
MANUFACTURING PROCESS
INSTRUCTIONS
Master Manufacturing
Formula
and
Master Manufacturing
Instructions
Manufacturing
Instructions
This section contains:
♦ Master Formula for 100 000 units (PIVOTAL)
♦ Formula comparison
♦ Equipment Comparison
Manufacturing Instructions
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
KEY:
Precautions: • Wear Mask and Gloves
‚ Wear disposable overalls
ƒ Use air stream face visor with AIR filter
„ Use Mask, Gloves and Safety glasses
Caution: …Avoid exposure to light / Protect form light
†Store in well closed containers
‡Potential danger to pregnant women
ˆPregnant women prohibited in this area
‰Do not store above 25øøC
• Tabletting Room humidity below 50%
6 Maximum time between granulation and compression
7 days.
y Pack blended granular material into a container with two
heavy duty plastic bags and seal 100% with a plastic tie.
> Place 2 x 500 g silica gel pack between plastic bags
ý 150 mg and 300 mg Ranitidine can not be geometrically
scaled.
Manufacturing Instructions
COMMERCIAL BATCH MASTER FORMULA
PAGE 1 of 3.
Per % RM. Sign
UNIT Exc Lot weigh.
ess Raw Material Names per 400 000 cores
mg No Dept.
Kg g mg L mL A B
PART ONE
168.0 - Ranitidine Hydrochloride Granulated 67 200
129.6 - Microcrystalline Cellulose NF 51 840
Avicel pH 102
9.60 - Croscarmellose Sodium NF 3 840
AC-DI-SOL
PART TWO
168.0 - Ranitidine Hydrochloride Granulated 67 200
-
PART THREE
4.80 - Magnesium Stearate NF 1 920
- -
480.0 - Theoretical End Weight. 192 000
PART FOUR AQUEOUS FILM COAT SUSPENSION ¹
10 Aqueous Film Coating Suspension
8.2 Methocel E-5 Premium 3 620
Hydroxypropyl Methylcellulose USP
3.41 OPASPRAY-K-7000 [White] 3 000
- Methyl Chloride NF 70 300
- Isopropyl Alcohol USP 46 780
Manufacturing Instructions
PIVOTAL BATCH MASTER FORMULA
Kg g mg L mL A B
PART ONE
168.0 - Ranitidine Hydrochloride Granulated 33 600
129.6 - Microcrystalline cellulose NF 25 920
Avicel pH 102
9.60 - Croscarmellose Sodium NF 1 920
AC-DI-SOL
PART TWO-
168.0 - Ranitidine Hydrochloride Granulated 33 600
-
PART THREE
4.80 - Magnesium Stearate NF 0 960
-
480.0 Theoretical End Weight. 96 000
- -
Manufacturing Instructions
COMMERCIAL BATCH MASTER FORMULA
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97
Kg g mg L mL A B
PART FOUR - FILM COATING,
SUBLOT 1
300.0 Ranitidine 150 mg CORES 96 000 [200 000 Cores]
Ranitidine 150 mg core Aqueous 61 850
6.0² Film Coating Suspension (3.95%
solids in COATING SUSPENSION)
306.0 Theoretical End Weight. 98 000
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
PAGE 1 of 5.
Machine Sign Date
MANUFACTURING INSTRUCTIONS No: A B
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97
5. LOAD into the [Y-Cone blender] (Type 200) with the MAGNESIUM
STEARATE:
6. Mix for 5 minutes.
Total Mixing Time is 5 minutes.
Start of Mixing Time - [ ] am /pm
Time of mixing - [ ] am /pm
7. PASS the BLENDED MATERIAL through VIBRATING ELECTRIC
SIEVE fitted with a 2.5 mm screen into a sufficient 100 liter containers or
bins lined with 2 heavy duty plastic bags. Place 2 x 500g bags of silica
gel drying agent between the bags.
8. WEIGH the milled granulate. __________Kg. Immediately add the
batch number to the scale print-out, and attach to the manufacturing
instructions, date and sign the print-out.
Manufacturing Instructions
Calculate Yield:
Yield ___________ %
10. TRANSFER the milled granulate from stage 8 to the compression
Department. - Maximum storage period - 7 days in sealed bags.
Date: ______ Time _________
11. COLLECT 10 samples, each equivalent to the approximate weight of
three tablet (900 mg) in labeled sample containers. Collect samples from
upper, middle and lower part of each the container.
Send the samples to the QC laboratory for Blend Uniformity Testing.
12. IDENTIFY and verify the cleanliness of the tabletting equipment and
de-duster in use.
COMPRESS the final blend according to the written product
specifications
Tabletting machine: Type & No KILIAN TX
Machine Speed: 100 000 cores per hour
Limit of Speed : NLT 80 000 ; NMT 120 000 cores per hour.
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
PAGE 3 of 5.
MANUFACTURING INSTRUCTIONS
Machine Sign Date
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97 A B
PART SIX
ORGANIC FILM COATING SOLUTION
15. IDENTIFY and verify the cleanliness of the coating equipment.
Weigh:
[ ] Kg METHYL CHLORIDE NF
[ ] Kg ISOPROPYL ALCOHOL USP
into a stainless steel vessel fitted with a ROLLER MIXER. [# ]
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
PAGE 4 of 5.
MANUFACTURING INSTRUCTIONS
Machine Sign Date
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97 A B
COATING PROCEDURE - SUBLOT ONE
Wear Masks and Gloves.
Caution : Inflammable Solvents.
Coating Procedure:
21. IDENTIFY and verify the cleanliness of the coating equipment in use
Sublot Size[ 96 ] Kg. Equal to [200 000] ___________ Caplets.
SUB LOT No 1.
PREHEATING OF CORES:
Coating machine (Type & No: ACCELA-COTA) :
Incoming Air Temperature: [50]º C - [55]º C
Total Warming time: [10] Minutes
Drum Speed: MINIMUM rpm (minimum speed)
Jogging cycle: One cycle every [ 2 ] minutes.
Extraction Air Temperature: [35]º C - [42]º C
Limit of rpm: NLT MINIMUM rpm:NMT MINIMUM rpm.
SPRAYING PARAMETERS:
Pump type: REXON AIRLESS No: 107802
Spray rate: [000] - [000] g/min
Nozzles: [ 2 ] - [ ] mm (No: 1365)
Angle of Guns to Bed: 90 º degrees
Height above Bed: [ 25 ] cm
Incoming Air Temperature: [ 50 ]º C - [ 55 ]º C (Target: 53 º C)
Extraction Air Temperature: [ 35 ]º C - [ 42 ]º C (Target: 38 º C)
22. COAT tablet cores at a drum speed of [ 5 ] rpm and a pump pressure
1.5 for 10 minutes and then at a drum speed of [ 6] rpm at 1.5-2.0 Atms.
Target COATED tablet weight of [ 486.5 ] mg is obtained.
COATING COMPLETION PROCEDURE
23. REDUCE drum speed to minimum rpm and perform the following:
(1) - Increase SET POINT of incoming air to 90ºC for 30 mins.
(2) - Check Relative Humidity (via ROTRONIC Equipment)
(3) - if RH % is less than 10 % go to sub-step (5) RECORD [ca 7%] RH
(4) - if RH is more than 10 % dry for an extra 30 min. CHECK RH %
again
(5) - cool until the Extraction Air Temperature reaches 45ºC - 50ºC
24. TRANSFER coated tablets into containers lined with two PE plastic
bags (clear inner, black outer) Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
25. ATTACH the temperature graphs (Type & No) to the manufacturing
instructions. Immediately add the batch / Sublot number to the
temperature graph(s) and date and sign graph.
26. WEIGH Coated tablets
Actual production weight: _______ [ ] Kg. No of Containers _____
Ed Number: 02 Effective Date:
APPROVED
Ed. Status: 01 DD/MM/YY P West E Hollmann C Frost C Latham R Ford
Department R &D RA QC / QA
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
PAGE 5 of 5.
MANUFACTURING INSTRUCTIONS
Machine Sign Date
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97 A B
COATING PROCEDURE - SUBLOT TWO
Wear Masks and Gloves.
Caution : Inflammable Solvents.
Coating Procedure:
21. IDENTIFY and verify the cleanliness of the coating equipment in use
Sublot Size[ 96 ] Kg. Equal to [200 000] ___________ Caplets.
SUB LOT No 1.
PREHEATING OF CORES:
Coating machine (Type & No: ACCELA-COTA) :
Incoming Air Temperature: [50]º C - [55]º C
Total Warming time: [10] Minutes
Drum Speed: MINIMUM rpm (minimum speed)
Jogging cycle: One cycle every [ 2 ] minutes.
Extraction Air Temperature: [35]º C - [42]º C
Limit of rpm: NLT MINIMUM rpm:NMT MINIMUM rpm.
SPRAYING PARAMETERS:
Pump type: REXON AIRLESS No: 107802
Spray rate: [000] - [000] g/min
Nozzles: [ 2 ] - [ ] mm (No: 1365)
Angle of Guns to Bed: 90 º degrees
Height above Bed: [ 25 ] cm
Incoming Air Temperature: [ 50 ]º C - [ 55 ]º C (Target: 53 º C)
Extraction Air Temperature: [ 35 ]º C - [ 42 ]º C (Target: 38 º C)
22. COAT tablet cores at a drum speed of [ 5 ] rpm and a pump pressure
1.5 for 10 minutes and then at a drum speed of [ 6] rpm at 1.5-2.0 Atms.
Target COATED tablet weight of [ 486.5 ] mg is obtained.
COATING COMPLETION PROCEDURE
23. REDUCE drum speed to minimum rpm and perform the following:
(1) - Increase SET POINT of incoming air to 90ºC for 30 mins.
(2) - Check Relative Humidity (via ROTRONIC Equipment)
(3) - if RH % is less than 10 % go to sub-step (5) RECORD [ca 7%] RH
(4) - if RH is more than 10 % dry for an extra 30 min. CHECK RH %
again
(5) - cool until the Extraction Air Temperature reaches 45ºC - 50ºC
24. TRANSFER coated tablets into containers lined with two PE plastic
bags (clear inner, black outer) Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
25. ATTACH the temperature graphs (Type & No) to the manufacturing
instructions. Immediately add the batch / Sublot number to the
temperature graph(s) and date and sign graph.
26. WEIGH Coated tablets
Actual production weight: _______ [ ] Kg. No of Containers _____
Ed Number: 02 Effective Date:
APPROVED
Ed. Status: 01 DD/MM/YY P West E Hollmann C Frost C Latham R Ford
Department R &D RA QC / QA
Manufacturing Instructions
COMMERCIAL BATCH MASTER FORMULA
OUTLINE OF
STANDARD OPERATING PROCEDURES FOR:
IN-PROCESS CONTROLS
STAGE 1.
STAGE 2.
STAGE 3.
STAGE 4.
T he Quality Assurance Unit reviews the batch test results and evaluates the
criteria of acceptance or rejection of each batch lot manufactured.
Manufacturing Instructions
Ranitidine HCl Tablets 300.0 mg Cat: 8602-97
KEY:
(1)
The testing frequency is performed twice when the overall compression running time is less
than four hours for the entire batch lot.
Deviations from the specifications and acceptance criteria, arising during the in-process
(2)
controls, shall determine the corrective/adjustment action performed on the tableting machinery
during the tablet compression stage.
The Double Limits for the Tablet Hardness Test are defined as C-±20% from the minimum
(3)
and maximum product specifications limits. When, there is a NLT 10 SCU Hardness
specification, the double limits may not exceed a minimum value of 8 SCU. (Not to go below 8
SCU).
Double Limits for Tablet Thickness Tests are defined as C ± 0.1mm from the minimum and
(4)
Manufacturing Instructions
COMMERCIAL BATCH MASTER FORMULA
Ranitidine HCl Caplets 300.0 mg Cat : 8613-97
Checks performed by Quality Control personnel
Quality Control personnel test the physical specifications of random samples
according to the individual product specifications sheets: A minimum sampling
frequency is tabulated for each eight hour (shift) period.
Quality Control - In-process Testing Schedule:
PAGE 2 of 2
Test Sample Frequency Acceptance
PERFORMED Size per shift (1) Criteria (2)
(Tablets) (min.)
INDIVIDUAL TABLET 20 (1) Twice NMT 2 tablets out of the 20 tested can
deviate from product spec. No deviation
WEIGHT
is allowed from Double Limits(3) spec.
THICKNESS 10 (1) Twice NMT 2 tablets out of the 10 tested can
deviate from product spec. No deviation
is allowed from Double Limits(4) spec.
HARDNESS 10 (1) Twice NMT 2 tablets out of the 10 tested can
deviate from product spec. No deviation
is allowed from Double Limits(5) spec.
DIAMETER Once No deviation from product specification
3 (1)
at start is allowed.
FRIABILITY 20 -40 (1) Twice
No deviation from product specification
According to
is allowed
product
DISINTEGRATION 6 (1) Twice No deviation from product specification
is allowed
KEY:
(1)Samples are taken, independently by QC personnel for batch release purposes, at least
once per hour throughout the tabletting run, producing a total representative sample quantity
of 300 -500 tablets. This representative sample lot is for QC batch release purposes .
(2)Deviations from specifications and acceptance criteria, arising during the in-process
controls, shall determine the corrective action to be performed on the tabletting machinery
during the compression stage.
(3) Double
Limits for the Individual Tablet Weight test are defined as the double value from the
minimum or maximum limit in relation to the nominal tablet value (i.e. target weight value).
Double Limits for Tablet Thickness Tests are defined as C - ± 0.1 mm from the minimum
(4)
Manufacturing Instructions
Ranitidine HCl Tablets 300.0 mg Cat: 8613-97
Quick Sieve
Sieve MESH # [50]
IPQC testing
STEP FOUR Blend Uniformity
VIBRATING SIEVING
2.5 mm
Yield Analysis
BLENDING Yield
Tabletting
Yield Analysis STEP FIVE
Compression stage IPQC testing
Tabletting Physical tests
Assay
Finished Product
FP TESTING
PHYSICAL TESTS
ASSAY
IMPURITIES / RH
DISSOLUTION
OVI
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
ATTACHMENTS:
Drying
N/A
Milled granulate
N/A
Final Blend
Attachment # 1 Mixing time Print-Out of the Final Blend
Attachment # 2 Weight Print-Out of the Final Blend
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
COMMERCIAL BATCH
PAGE 1 of 4
Total Final Blend Yield Limit: NLT 98.0% (based on actual quantities
processed).
In-Process
Final Blend Uniformity Limit: 94.0 - 106.0% of labeled amount
RSD ≤ 6.0% (as per attached specifications)
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
COMMERCIAL BATCH
PAGE 2 of 4
Individual core weight (±7.5%) Nominal 480.0 Limit: 444.0 - 516.0 mg:
Average core weight (±5.0%) Nominal 480.0 Limit: 456.0 -504.0 mg:
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
COMMERCIAL BATCH
PAGE 3 of 4
Product: Generic name Ranitidine HCl Caplets 300.0 mg.. Cat: 8613-97
Grams Grams
Film Coating Controls Before Coating After Coating
¹TARGET Tablet weight (mg) [480.0] [486.0]
CHECK WEIGHTS
¹Target Coated weight (mg) - [30.250]
In-Process Yields
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
Product: Generic name Ranitidine HCl Caplets 300.0 mg. Cat: 8613-97
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
[Avicel pH 102]
[AC-DI-SOL[
2. The actual quantity of a non active ingredient such as [Excipient NF] used in the formula will depend on
the WEIGHT for [Active Salt] used.
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
Caplets
PAGE 1 of 1
Equipment and Executed Batch Production
Manufacturing Conditions 100,000 units Batch
200,000 units
Premixing Ê Ê
Granulating - (High Shear Granulator) Ê Ê
FBD Drying Ê Ê
Sieve - Oscillating ü ü
Blending Y-cone 200 Y-cone 200
Manufacturing Instructions
MANUFACTURING INSTRUCTIONS FOR COMMERCIAL PRODUCTION
Stage One.
PACKAGING COMPONENTS:
1. Bulk Product
2. HDPE Containers
3. Package Insert or Outsert (Product Leaflets)
4. Desiccant (Silica Gel)
5. Container Label
6. Master Cartons
7. Carton Shipping Labels
Stage Two
PACKAGING PROCEDURE:
Manufacturing Instructions
PAGE 1 of 1
(1) Average figures for containers per minute output for Slow and High Speed.
(2) All indicated machine outputs are adjusted to the Tablet Slate Filler rate.
TABLE OF CONTENTS
(Overall ANDA Guideline Requirements for this Section).
◊ Formula comparison
◊ Equipment Comparison
◊ Reprocessing Statement
24 Volume
V Drug Development Series: Sect: 7.45 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.46 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
ENTERIC COATED
TABLETS
DICLOFENAC Na EC TABLETS
50.0 & 75.0 mg.
ALCOHOLIC GRANULATION
24 Volume
V Drug Development Series: Sect: 7.47 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.48 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 1 of 2 pages.
Per % Raw Material Names RM. Sign
UNIT Ex Lot weigh.
ce For [1000 000] kg
mg No Dept.
ss
Kg G mg
PART ONE
50.0 Diclofenac Sodium 50 000
90.0 Lactose Monohydrate NF (200 90 000
mesh)
7.6 Povidone USP (PVP K-30) 7 600
10.0 Sodium Starch Glycolate NF 10 000
12.4 Starch NF 12 400
PART TWO
GRANULATION SOLUTION
- Alcohol USP 25 000
- Alcohol USP q.s. (up to 4.2 kg) 000 000
PART THREE
18.5 Microcrystalline Cellulose NF 18 500
(Avicel PH-101
0.5 Aerosil 200 0 500
(Colloidal Silicon Dioxide NF)
PART FOUR
1.0 Magnesium Stearate NF 1 000
TOTAL 190 000
190.0 DICLOFENAC SODIUM E.C. 190 000
TABLETS 50 mg CORES
* 13.6 DICLOFENAC SODIUM TABLETS 50 109 500
mg FILM COATING DISPERSION
203.6 TOTAL (Theoretical) 203 600
Ed Number:
01
Effective Date:
APPROVED
Ed. Status DD/MM/YY _____________ __________ _______________ _________/________
New Department R &D RA QC / QA
* Average solids remaining on the tablet core
24 Volume
V Drug Development Series: Sect: 7.49 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 2 of 2 pages.
Per % RM. Sign
Ex
Raw Materials
UNIT Lot For [1000 000] kg weigh.
mg ce No Dept.
ss
Kg G mg
20 ENTERIC COATING
SUSPENSION
PART ONE
0.88 20 Polyethylene glycol 6000 NF 1 050
- Purified water USP (60°C) 5 400
PART TWO
0.88 Talc USP 1 060
0.63 Titanium Dioxide USP 0 760
0.1 Color Ferric Oxide NF Red 0 120
0.05 Color Ferric Oxide NF Yellow 0 060
- Purified water USP 14 200
PART THREE
11.121 Methacrylic Acid Copolymer NF 44 480 (Ref)
(Eudragit L30D-55 SOLUTION
33%)
- Purified water USP 54 000
PART FOUR
13.66 TOTAL 163 900
190.0 DICLOFENAC SODIUM E.C. 190 000
TABLETS 50 mg CORES
* 13.6 DICLOFENAC SODIUM TABLETS 50 109 500
mg FILM COATING DISPERSION
203.6 TOTAL (Theoretical) 203 600
1
Solids (amount of solids = 30% of
the solution)
Ed Number:
01
Effective Date:
APPROVED
Ed. Status DD/MM/YY _____________ __________ _______________ _________/________
New Department R &D RA QC / QA
(Ref. calculation) 11.2 (solids) x 1.2 (20% excess) / 0.3 (solution concentration) = 44.480 Kg = (Weight of solution)
24 Volume
V Drug Development Series: Sect: 7.50 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 1 of 2 page.
Per % Raw Material Names RM. Sign
UNIT Ex Lot weigh.
ce For [1000 000] kg
mg No Dept.
ss
Kg G mg
PART ONE
75.0 Diclofenac Sodium 75 000
135.0 Lactose Monohydrate NF (200 135 000
mesh)
11.35 Povidone USP (PVP K-30) 11 350
15.0 Sodium Starch Glycolate NF 15 000
18.5 Starch NF 18 500
PART TWO
GRANULATION SOLUTION
- Alcohol USP 25 000
- Alcohol USP q.s. (up to 4.2 kg) 000 000
PART THREE
28.15 Microcrystalline Cellulose NF 28 150
(Avicel PH-101
0.5 Aerosil 200 0 500
(Colloidal Silicon Dioxide NF)
PART FOUR
1.5 Magnesium Stearate NF 1 500
TOTAL 285 000
285.0 DICLOFENAC SODIUM E.C. 285 000
TABLETS 75 mg CORES
21.6 DICLOFENAC SODIUM TABLETS 75 173 500
mg ENTERIC COATING DISPERSION
306.6 TOTAL (Theoretical) 306 600
Ed Number:
01
Effective Date:
APPROVED
Ed. Status DD/MM/YY _____________ __________ _______________ _________/________
New Department R &D RA QC / QA
24 Volume
V Drug Development Series: Sect: 7.51 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 2 of 2 page.
Per % RM. Sign
Ex
Raw Materials
UNIT Lot For [1000 000] kg weigh.
mg ce No Dept.
ss
20 ENTERIC COATING Kg G mg
SUSPENSION
PART ONE
0.99 20 Polyethylene glycol 6000 NF 1 190
1.62 Purified water USP (60°C) 5 100
PART TWO
0.99 Talc USP 1 190
0.89 Titanium Dioxide USP 1 065
0.033 Color Ferric Oxide NF Red 0 040
0.017 Color Ferric Oxide NF Yellow 0 020
2.92 Purified water USP 14 100
PART THREE
12.431 Methacrylic Acid Copolymer NF 49 720 (Ref)
(Eudragit L30D-55 SOLUTION
33%)
0.82 Purified water USP 50 600
21.6 TOTAL 163 900
PART FOUR
2
Residual moisture after film drying of enteric coat.
24 Volume
V Drug Development Series: Sect: 7.52 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.53 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.54 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Stage 19. Store the GRANULATE at NMT 25C until required for
use within 10 days.
Stage 20.
Compression
Verify cleanliness / Identification of tableting equip.
Identify and verify the cleanliness of the tablet equipment in use
COMPRESS according to the written product specifications
Stage 21.
Tablet machine: (Type & No [KILIAN TX / FETTA).
Machine Speed 90 000 Tablets per hour
Limit of rpm NLT 80 000 tph ; NMT 100 000 tph
Stage 22.
Weigh the Tablet CORE:
(Average weight of a Tablet core: - (190 mg)
Actual production weight: Ü [000.0] Kg.
Weight of Samples taken: less [00.0] Kg.
Vacuum and rejects Weight: less [00.0] Kg.
Total overall weight = [000.0] Kg
No of Bulk Containers filled = [0]
Theoretical Weight [190] Kg. Yield:- [00.0]%
(Yield Limits: NMT 2% unexplained loss compared to the final
blend weight from stage 15.
Stage 23.
Seal the double PE plastic bags (clear inner, 2 silica 500g bags,
black outer) with plastic ties then close all containers, and attach
(bar coded) labels to the Bulk Containers for transport to the
holding area PRIOR TO THE ENTERIC COATING PROCESS.
24 Volume
V Drug Development Series: Sect: 7.55 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.56 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 5 of 5 pages
Coating Procedure:
Stage 31. IDENTIFY and verify the cleanliness of the coating equipment
in use
Sublot Size [95.0] Kg. Equal to [500 000] tablets.
PREHEATING OF CORES:
Coating machine (Type & No) : [[ACCELA COTA AC150 / AC48 ]]
Extraction Air Temperature: [[29]º C - [[37]]º C
Incoming Air Temperature: [48]º C - [[56]] º C
Total Warming time: [2] Minutes
Drum Speed: [2] rpm (minimum speed)
Jogging cycle: One cycle every [2] minutes.
Limit of rpm: NLT [2] rpm: MT [3] rpm.
SPRAYING PARAMETERS
Pump type: Peristaltic
Spray rate: [200] - [700] g/min
Nozzles: [[3]] x [[1.5] mm
Angle of Guns to Bed: 90 degrees
Height above Bed: [25] cm
Incoming Air Temperature: [[48]]º C - [[56]]º C (Target: [50]ºC)
Extraction Air Temperature: [[29]]º C - [[37]]º C (Target: [32]ºC)
Stage 32. COAT tablet cores at a drum speed of [[3]] - [[9]] rpm and a
spray rate of [200] -[700] g/min until the target COATED tablet weight
of [203.6] for the 50 mg core and [306.6] mg for the 75 mg core is
obtained. Monitor the weight gain at 10-15 min intervals and record the
results obtained
COMPLETION OF COATING PROCEDURE
Stage 33. REDUCE drum speed to minimum rpm and perform the
following:
n Reduce set point of incoming air temperature to [40] ºC
n on reaching this temperature - close the inlet air
n continue drum speed until the Extraction Air Temperature reaches
[26] ºC - [30]ºC
Stage 34. TRANSFER coated tablets into containers lined with two PE
plastic bags (clear inner, black outer) Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
Stage 35. ATTACH the temperature graphs (Type & No) to the
manufacturing instructions. Immediately add the batch / Sublot number to
the temperature graph(s) and date and sign graph.
Stage 36. WEIGH EC Coated tablets
Actual production weight: [000] Kg. No of Containers [0]
24 Volume
V Drug Development Series: Sect: 7.57 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.58 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Manufacturing Instructions
DICLOFENAC Na EC TABLETS 75.0 mg. Lot: IA000-00
Page 2 of 5 pages
MANUFACTURING INSTRUCTIONS Machine Sign Date
for 75 mg tablets
PART ONE - PREPARATION OF GRANULES
Stage 7. Grind by Frewitt oscillating granulator a small
quantity of the granulate of stage # 6 through 1.0 mm screen
Stage 8. Check Loss on Drying of the grounded granulate
(using Computrac-Max. 50, temperature 110°C
LOD required = 1.0 - 2.0%
LOD Result = ________%
If necessary, continue to dry the granulate until the required
LOD is obtained. Final LOD Result = ________%
Stage 9. Grind the dried granulate by Frewitt oscillating
granulator through 1.0 mm screen.
Stage 10. WEIGH the milled granulate.
Stage 11. SIEVE through a 30 mesh screen or quick sieve #
1.5 mm. ADD the,
MICROCRYSTALLINE CELLULOSE NF (AVICEL PH-101)
Stage 12. TRANSFER the granulate and the sieved powder
of stage 11 into Y-Cone 500 and mix for 15 minutes
Stage 13. SIEVE MAGNESIUM STEARATE NF through a 30
mesh screen.
Stage 14. ADD the screened MAGNESIUM STEARATE NF
from stage # 13 to the mixture of stage #12 and mix for an
additional 5 minutes [max.]
Stage 15. PASS the lubricated granulate from stage #14
through a Quick Sieve 3 mm screen into a [FLOW] BIN
Stage 16. COLLECT 10 samples from the lower, middle and
upper part of the 500 L CONTAINER/ [FLOW] BIN.
[1]. Weight of each sample is equivalent the fill weight of
NMT Three Tablets 75 mg.
[2]. Forward samples to the analytical laboratory for BLEND
UNIFORMITY testing.
Stage 17. Immediately add the batch number to the scale
print-out, attach to the manufacturing instructions after dating
and signing it.
Ed Number: Effective Date:
01
APPROVED
Ed. Status DD/MM/YY _____________ __________ _______________ _________/________
New Department R &D RA QC / QA
24 Volume
V Drug Development Series: Sect: 7.59 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Manufacturing Instructions
DICLOFENAC Na EC TABLETS 75.0 mg. Lot: IA000-00
Page 3 of 5 pages
MANUFACTURING INSTRUCTIONS Machine Sign Date
for 75 mg tablets
Stage 18. Seal the blended material in extra heavy duty double
PE plastic bags (clear inner, 2 silica 500g bags, black outer) with
plastic ties then close all containers, and attach (bar coded) labels
to the Bulk Containers for transport to the holding area
Theoretical Weight [000.0] Kg. Yield [00.0] %
Stage 19. Store the granulate at NMT 25C until required for use
within 10 days.
Stage 20.
Compression
Verify cleanliness / Identification of tableting equip.
Identify and verify the cleanliness of the tablet equipment in use
COMPRESS according to the written product specifications
Stage 21.
Tablet machine: (Type & No [KILIAN TX / MANESTY/ FETTA).
Machine Speed 90 000 Tablets per hour
Limit of rpm NLT 80 000 tph ; NMT 100 000 tph
Stage 22.
Weigh the tablet cores:
(Average weight of a tablet core:- (285 mg)
Actual production weight: Ü [000.0] Kg.
Weight of Samples taken: less [00.0] Kg.
Vacuum and rejects Weight: less [00.0] Kg.
Total overall weight = [000.0] Kg
No of Bulk Containers filled = [0]
Theoretical Weight [285] Kg. Yield:- [00.0]%
Equivalent to tablet cores [1 000 000.0]
(Yield Limits: NMT 2% unexplained loss compared to the final
blend weight from stage 15.
Stage 23.
Seal the double PE plastic bags (clear inner, 2 silica 500g bags,
black outer) with plastic ties then close all containers, and attach
(bar coded) labels to the Bulk Containers for transport to the
holding area.
24 Volume
V Drug Development Series: Sect: 7.60 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.61 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Page 5 of 5 pages
Coating Procedure:
Stage 31. IDENTIFY and verify the cleanliness of the coating equipment
in use
Sublot Size [142.5] Kg. Equal to [500 000] tablets.
PREHEATING OF CORES:
Coating machine (Type & No) : [[ACCELA COTA AC150 / AC48 ]]
Extraction Air Temperature: [[29]º C - [[37]]º C
Incoming Air Temperature: [48]º C - [[56]] º C
Total Warming time: [2] Minutes
Drum Speed: [2] rpm (minimum speed)
Jogging cycle: One cycle every [2] minutes.
Limit of rpm: NLT [2] rpm: MT [3] rpm.
SPRAYING PARAMETERS
Pump type: Peristaltic
Spray rate: [200] - [700] g/min
Nozzles: [[3]] x [[1.5] mm
Angle of Guns to Bed: 90 degrees
Height above Bed: [25] cm
Incoming Air Temperature: [[48]]º C - [[56]]º C (Target: [50]ºC)
Extraction Air Temperature: [[29]]º C - [[37]]º C (Target: [32]ºC)
Stage 32. COAT tablet cores at a drum speed of [[3]] - [[9]] rpm and a
spray rate of [200] -[700] g/min until the target COATED tablet weight
of [203.6] for the 50 mg core and [306.6] mg for the 75 mg core is
obtained. Monitor the weight gain at 10-15 min intervals and record the
results obtained
COMPLETION OF COATING PROCEDURE
Stage 33. REDUCE drum speed to minimum rpm and perform the
following:
n Reduce set point of incoming air temperature to [40] ºC
n on reaching this temperature - close the inlet air
n continue drum speed until the Extraction Air Temperature reaches
[26] ºC - [30]ºC
Stage 34. TRANSFER coated tablets into containers lined with two PE
plastic bags (clear inner, black outer) Seal bags with plastic ties and close
containers, and attach (bar coded) labels to the Bulk Containers for
transport to the holding area.
Stage 35. ATTACH the temperature graphs (Type & No) to the
manufacturing instructions. Immediately add the batch / Sublot number to
the temperature graph(s) and date and sign graph.
Stage 36. WEIGH EC Coated tablets
Actual production weight: [000] Kg. No of Containers [0]
24 Volume
V Drug Development Series: Sect: 7.62 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
FLOW CHART
SUBLOT #1 SUBLOT #2
24 Volume
V Drug Development Series: Sect: 7.63 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
FLOW CHART
SUBLOT #1 SUBLOT #2
FILM COATING SUSPENSION FILM COATING SUSPENSION
24 Volume
V Drug Development Series: Sect: 7.64 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
ATTACHMENTS:
THE FOLLOWING ATTACHMENTS ARE PLACED HERE:
Mixing Time
Attachment # 1 Mixing time Print-Out - Step 2
Attachment # 2 Mixing time Print-Out - Step 3
Attachment # 3 Mixing time Print-Out - Step 4
DRYING Temp
Attachment # 4 FDB Temperature/Time Graph - Step 6
Attachment # 5 Milled granulate LOD Print-Out - Step 8
Final Blend
Attachment # 6 Weight Print-Out of the milled granulate - Step
10
Attachment # 7 Mixing time Print-Out of the Intermediate Blend - Step 12
Attachment # 8 Mixing time Print-Out of the Final Blended material - Step 14
Attachment # 9 Weight Print-Out of the Final Lubricated material - Step 16/18
Compressed Tablets
Attachment # 10 Weight Print-Out of the bulk tablets - Step 22
Coated Tablets
Attachment # 11 FDB Temperature/Time Graph - Step 31
Attachment # 12 Weight Print-Out of the bulk tablets - Step 32
24 Volume
V Drug Development Series: Sect: 7.65 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
EXECUTED BATCH
Yields
Final Blend Yield Limit: NLT 98.0%
Total Final Blend Yield Limit: NLT 98.0% (based on actual quantities
processed).
In-Process
Final Blend Uniformity Limit: 94.0 - 106.0% of labeled amount
RSD ≤ 6.0% (as per attached specifications)
Blend Uniformity
The requirements for Blend Uniformity are met if the amount of the active ingredient in each
of the 10 samples, as determined from the Blend Uniformity Analytical Method, lies within
the range of 90.0 - 110.0% of the labeled amount and the Relative Standard Deviation is
less than or equal to 6.0%.
If 1 sample is outside the range of 90.0 - 110.0% of labeled amount and no sample is
outside the range of 80.0 - 120.0% of labeled amount, or if the Relative Standard Deviation
is greater than 6.0%, or if both conditions prevail, test 20 additional samples.
The requirements are met if not more than 1 sample of the 30 is outside the range of 90.0 -
110.0% of labeled amount and no sample is outside the range of 80.0 - 120.0% of labeled
amount, the Relative Standard Deviation of the 30 samples does not exceed 7.8%.
24 Volume
V Drug Development Series: Sect: 7.66 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
SUMMARY
Product: DICLOFENAC Na CORES 50 - 75 mg LOT: IA000-00
Labeled Amount: Each tablet contains DICLOFENAC Na [50.0]; [75.0 mg].
In-process tablet specifications
Punch Diameter 50.0mg 8.4 mm Punch No [P044] / Die No [D046]
Punch Diameter 75.0mg 9.5 mm Punch No [P058] / Die No [D058]
Description White round biconvex CORES debossed
with the number [50 or 75] on one face of the
core
Scoring [not scored]
Core Diameter 50mg Nominal 8.4 mm Limit: 8.2 - 8.8 mm
Core Diameter 75mg Nominal 9.5 mm Limit: 9.4 - 9.8 mm
Disintegration Time NMT 15 min
Method - USP Medium - Purified Water USP
50 mg TABLET
Individual core weight (±7.5%) Nominal 190.0 Limit: 176.0 - 204.0 mg
Average core weight (±5.0%) Nominal 190.0 Limit: 180.5 - 199.0 mg
75 mg TABLET
Individual weight (±7.5%) Nominal 285.0 Limit: 263.6 - 306.3 mg:
Average weight (±5.0%) Nominal 285.0 Limit: 270.8 - 299.0 mg:
Thickness 50.0mg Nominal 2.5 Limit: 3.0 - 4.00 mm
Thickness 75.0mg Nominal 4.2 Limit: 3.7 - 5.00 mm
24 Volume
V Drug Development Series: Sect: 7.67 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
EXECUTED BATCH
In-Process Yields
24 Volume
V Drug Development Series: Sect: 7.68 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
24 Volume
V Drug Development Series: Sect: 7.69 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
COMPRESSION
TABLET Machine KILIAN TX KILIAN TX
MANESTY MANESTY
PACKAGING
Packaging Units King / Lakso King / Lakso
Labeling Groninger Groninger
Prestek Prestek
FACILITIES
Equipment Variation NONE NONE
Manufacturing Area Production Production
Staff Production Production
SOP Production Production
24 Volume
V Drug Development Series: Sect: 7.70 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
DICLOFENAC Na Tablets
50 & 75 mg
AC-150 AC-150
24 Volume
V Drug Development Series: Sect: 7.71 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
1
Processing Solvent only.
2
Solids (amount of solids = 30% of the solution)
3
Including 20% excess
24 Volume
V Drug Development Series: Sect: 7.72 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
1
Processing Solvent only.
2
Solids (amount of solids = 30% of the solution)
3
Including 20% excess
24 Volume
V Drug Development Series: Sect: 7.73 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
Stage Two
PACKAGING PROCEDURE:
HDPE Container & Bulk Line Feed
Cotton Coil
24 Volume
V Drug Development Series: Sect: 7.74 Oral DR TABLETS
HANDBOOK of GENERIC DRUG DEVELOPMENT ANDA DEVELOPMENT
(1) Average figures for containers per minute output for Slow and High Speed.
All indicated machine outputs are adjusted to the Tablet Slate Filler rate.
(2)
24 Volume
V Drug Development Series: Sect: 7.75 Oral DR TABLETS
ORAL TABLETS IN-PROCESS CONTROLS CHAPTER 8
production batch.
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
1. All dry powders are screened (state mesh size) prior to processing? qYes qNo
2. Mixing of small weight excipients in plastic bags is not permitted ? qYes qNo
3. The granulating agent is dissolved in the granulating solvent (at a qYes qNo
specific temperature) with a clearly defined mixing time?
5. The rate of addition for critical ingredients (e.g. spraying of granulating qYes qNo
solution) is clearly identified and documented in the manufacturing
procedure
6. Compatible excipients are grouped into a single manufacturing step qYes qNo
where suitable?
7. Granulating mixing times are identified and recorded by ‘start and qYes qNo
stop’ entries?
8. Granulating mixing speeds (rpm) and blade settings are identified ? qYes qNo
9. FBD inlet, outlet and bed temperatures and tray dryers operating qYes qNo
temperatures (±20 C) are identified as well as overall drying times.
10. Granulate moisture is controlled during drying stage as a critical qYes qNo
processing parameter (moisture frequently affects hardness and
dissolution during product aging).
11. The granulate is dried to a specific granulate moisture content by qYes qNo
milling a sample portion and evaluating the LOD using IR equipment ?
12. Drying is continued until the desired moisture content is obtained? qYes qNo
13. Milling speeds (rpm) sieve size (rpm) and knife settings are qYes qNo
identified ?
Footnote : The procedure for selecting approved suppliers for non active ingredients will minimize
inter-batch excipient variation significantly. This variation, if present, may impact upon the in-process
controls. The absence of adequate GMP (cleaning procedures) may adversely affect the microbial
bioburden of the bulk material during the manufacture and the filling process.
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
Routine in-process quality controls on the final product consists of testing the bulk granulate
and the in-process tablets or cores during the compression stage: The following parameters
should be evaluated in the granulate, compressed tablets, or tablet cores and coated tablets:
14. Maximum processing times are stipulated for key stages and the qYes qNo
overall manufacturing time is controlled and documented ?
15. Routine IPQC. testing on the granulate material is controlled by: qYes qNo
⇒ moisture control (IR-LOD) on milled sample (at the drying stage) qYes qNo
16. Infra-red moisture testing unit for granulates is calibrated against qYes qNo
oven LOD moisture values.
17. LOD moisture values for granulates qualified by manufacturing qYes qNo
(drying) the product at the lower and upper limits of the Intermediate
Product Release Specification during the product development stage.
18. Infra-red moisture testing unit temperature setting determined. qYes qNo
19. Infra-red moisture testing unit type and model number qYes qNo
documented.
20. NB. A standard granulate quantity and uniform spreading on IR qYes qNo
trays is documented and emphasized in the IR - LOD method ?
21. Hardness Range Qualification - Range limits (e.g. hardness) have qYes qNo
been suitably qualified by manufacturing the product at the lower and
upper limits of the Finished Product Release Specification during the
product development stage (thickness, disintegration and dissolution
were evaluated) ?
22. In-process limits of tablet weight and hardness are set marginally qYes qNo
tighter than the product release specifications?
23. Target fill weight and rpm range limits for the tabletting procedure qYes qNo
were established during the product development stage and then
validated as a critical process parameter ?
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
24. Routine In-Process Quality Control testing performed on tablet qYes qNo
cores or uncoated tablets are:
25. Routine In-Process Quality Control testing on coated tablets are: qYes qNo
• Final Coated Weight (10 units) approximately 15 mg film coating qYes qNo
for a 18 x 8 mm caplet
• Final Coated Thickness (increase tablet thickness range from core to qYes qNo
coated - upper specification only, by ~0.5 - 1.0 mm)
Setting the limits for tablet thickness may be achieved by compressing the tablet at the extreme ends of the
hardness range (lowest and highest values) and measuring the resulting thickness.
The range may be skewed to the right, if necessary. As an example a 4.0 mm tablet - is set a range of 90 to
115% if there is a possibility of a significant variation in raw material bulk density (i.e. 3.6 to 4.6 mm).
The range specification may be reviewed in the forthcoming annual report after several batches have been
produced commercially.
Ø IN-PROCESS CONTROLS ×
CL # P-HGD-03-01Y2K
STAGE 2.
Appropriate in-process controls include material testing by the Quality Control Unit
and Quality Assurance personnel.
These test are:
⇒ Content uniformity of final blend.
⇒ Physical specifications of the Tablets.
STAGE 3.
In-process material testing is performed by responsible on-line manufacturing
personnel and qualified Quality Assurance Unit Personnel.
STAGE 4.
The Quality Assurance Unit reviews the batch test results and evaluates the criteria
of acceptance or rejection of each batch lot manufactured.
Ø IN-PROCESS CONTROLS ×
CL # HPGD-03-069Y
Ø IN-PROCESS CONTROLS ×
CL # HPGD-03-01Y2K
Deviations from specifications and acceptance criteria, arising during the in-process controls, shall
(2)
determine the corrective action to be performed on the tabletting machinery during the compression
stage.
Double Limits for the Individual Tablet Weight test are defined as the double value from the
(3)
minimum or maximum limit in relation to the nominal tablet value (i.e. target weight value).
(4)
Double Limits for Tablet Thickness Tests are defined as C - ± 0.1 mm from the minimum and
maximum specification limit values. Where C = limit value.
Double Limits for Tablet Hardness Test are defined as C-±20% from the minimum and maximum
(5)
product specifications limits. When, there is a NLT 10 SCU Hardness
specification, the double limits may not exceed a minimum value of 8 SCU. (i.e. not permitted to go
below 8 SCU).
Ø IN-PROCESS CONTROLS ×
IN-PROCESS CONTROL SPECIFICATION
GRANULATION MATERIAL
YIELD VALUES
Page 1 of 2.
Total Final Blend Yield Limit: NLT 98.0% (based on actual quantities
processed).
In-Process
Final Blend Uniformity Limit: 94.0 - 106.0% of labeled amount
RSD ≤ 6.0% (as per attached specifications)
Tabletting Yield
NMT 2.0% unexplained loss from the
previous final blend step.
Ø IN-PROCESS CONTROLS×
CORE CONTROLS
IN-PROCESS SPECIFICATIONS
Product: Tablets [USP] [000.0] mg.
Labeled Amount: Each tablet contains [000.0] mg [Active Material]
Caplet Debossing
Face ABC 72
Obverse XYZ
Ø IN-PROCESS CONTROLS×
IN-PROCESS CONTROL SPECIFICATION
ON COATED TABLET / CAPLET
PRODUCTION YIELDS
STANDARD OPERATING
PROCEDURES
CL # HPGD-03-01Y2K
4
[End of Document]
RELEASE SPECIFICATION
TABLET / CAPLET - COATED
Page 1 of 2.
Note: Residual Solvent / OVIs, if present in the film coat require a release specification.
1 Stability Check Specifications Tests performed during stability evaluation.
STABILITY SPECIFICATION
TABLET / CAPLET
Page 2 of 2.
PHYSICAL TESTS
NOTE:
Difference between release assay 95 - 105% and stability check assay 90.0 - 110.0
1Stability Check Specifications Tests recommended - if deemed to be necessary during product
development - (JUNE 1998 DRAFT STABILITY GUIDANCE TO INDUSTRY).
ØC H E C K L I S T ×
SOP # P-HGD-03-0Y2K
1. The Finished Product Specifications have both release and a stability qYes qNo
check specifications that allows for appropriate product aging throughout
the allocated shelf life ?
2. All the stability specifications are shown to be - stability indicating ? qYes qNo
3. Release specifications have narrower limits than the stability check qYes qNo
specifications, allowing an appropriate margin of safety as the product
ages ?
4. Development and qualification lots show content uniformity is similar qYes qNo
for the granulation material and the compressed tablets or uncoated
tablet cores ?
5. The firm performs specified critical in-process controls to insure that qYes qNo
the finished product testing is always in specification (e.g. content
uniformity) ?
7. The assay release specifications are set at 95.0 - 105.0% ? qYes qNo
8. The assay check specifications are set at 90.0 - 110.0% ? qYes qNo
9. Batches released at <97.0% are investigated and monitored for qYes qNo
stability, if development studies show active loss is >7% for claimed
shelf-life) ?
ØC H E C K L I S T ×
SOP # P-HGD-03-01Y2K
10. Tablet Hardness, Friability and Disintegration tests are not qYes qNo
performed as a finished product release test as these tests are
routinely addressed during in-process controls?
12. Tablet Hardness Range Qualification was performed to verify qYes qNo
that all the Check Finished Product Specification remained in
specification when processed at the lower and upper hardness
limit?
13. The Dissolution Test USP was evaluated in the development qYes qNo
and qualification batches and consistently demonstrated in the
pivotal and three full size commercial validation lots?
15. The Description allows for possible minor changes in color qYes qNo
from product release to end of shelf life (i.e. white to off-white
tablet)?
Footnote : Where the product batch history shows test failures due to environmental or raw material
variations is a clear indication that both these parameters should be further investigated and
addressed
STANDARD OPERATING
PROCEDURES
SOP # P-HGD-03-01Y2K Page 1 of 1
4
[End of Document]
Process
Optimization
‘…choosing the right formula and process specifications
prior to qualification…'
FORMULA FORMULA
FOR FOR
OPTIMISING OPTIMISING
LOD RANGE LUBRICANTS
Table No. 09
Tablet
Physical Parameters
Placed on
Stability &
tested for:- Physical
Assay Parameters
Impurities evaluated
Dissolution
Table No. 11
Assay and Impurities (%)
Batch P-07 - Containing BHA 0.10% w/w
Parameters Thin Layer (TLC) HPLC
Storage Appear BHAAssay Impurity Impurity Impurity ANY TOTAL Impurity ANY TOTAL
(months) ance % I II III Impurity I II Impurity Impurity
TLC TLC TLC I HPLC II II
Specs White to NMT 90.0 - NMT NMT NMT NMT NMT NMT NMT NMT
Off 50- 110.0 1.0 0.5 0.5 0.1 3.0 0.5 0.1 1.0
White 110
0 Conforms 90 102.2 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 <0.02 <0.04
1 Conforms 70 100.4 <0.1 <0.1 <0.1 <0.1 <0.3 <0.02 <0.02 <0.04
2 Conforms 70 102.0 <0.1 <0.1 <0.1 <0.1 <0.3 0.02 0.03 0.05
3 Conforms 60 101.8 <0.1 0.1 0.1 0.1 0.3 0.02 0.05 <0.07
Key:
Assay % of Label Claim of Active BHA Butylated Hydroxyanisole NF Dissolution % of Label Claim of Active
ANY I = Any other impurities (by TLC) TOTAL I = Total impurities (by TLC) ANY II = Any other impurities (HPLC)
TOTAL II = Total impurities (by HPLC)
Container / closure system : HDPE container 110cc Fill size: 150 Tablets
Storage conditions : 40°C / 75% Relative Humidity Mfg. Date:
Packaging Date : Expiration: Date Stability Start Date:
Table No. 12.
Storage Appearance
Assay & Dissolution
(%)
in Batchð
P-06 P-07
months Assayò 10’ 20’ 30’ 10’ 20’ 30’
Specs. White to Off White 90.0-110.0 NLT 75% (Q) NLT 75% (Q)
dissolved in 30’ dissolved in 30’
(2.1) (2.1)
(2.2) (2.2)
3
Conforms 100.5 96.1 97.2 98.7 97.1 97.2 96.7
(3.1) (2.6) (2.4) (2.0) (2.4) (2.4)
OPTIMUM
LOD & Lower &
LUBRICANT Upper LOD
COMBINATION. Range fully
qualified
DISSOLUTION
is in Specification
S C A L E - UP
P r ocedures
‘…Scale-up is a development procedure -
pivotal and validation lots are demonstration procedures…’
APPLICATION RATE O
2 C) for ~8 minutes and then with
The rate of coating solution delivery ambient air for an additional ~5
is an important process control minutes.
variable. Pre-warming the cores Discharge the coated tablets into a
allows for an initial fast coat of the double polyethylene lined drums, seal
primary layer. While fast application liners and drum and weigh. Record net
of the coating solution is important to weight and calculate yield.
minimize batch times, it must be Scale-up coating problems
remembered that there are Tablet Coats may slow down the
limitations to each type of equipment dissolution during the initial 10-15
and coating solutions being utilized. minute sampling. PVP K#30 tablet
Practical limitations can be binder in granulating solutions (at 2-
determined by utilizing the basic 4%) and certain commercial HPMC
thermodynamic relationships and coating solutions may slow down the
monitoring exhaust air temperature. initial curve of the dissolution profile,
It is important to make the smallest especially accelerated stability
o
possible droplet size to insure rapid samples (40 C/75%RH).
drying. Pigmented coating problems
The amount of air being applied and Tablet Coats are frequently opaque
the amount of pressure being utilized pigmented coatings. Clear
to atomize the liquid droplets can transparent coats are occasionally
determine the efficiency and used. Pigmented coating can provide
effectiveness of the coating system. additional light stability to dosage
Air atomization is generally preferred forms and help to differentiate tablets
with aqueous systems as it enhances by color coding. Pigments used in
the initial liquid droplet evaporation. tablet coating are generally
Small droplets are necessary to aluminum lakes or iron oxides with
achieve a fine smooth surface on titanium dioxide or talc used in white
coating tablets. coats or for color dilution in pastel
The flow rate of the coating solution colors.
is generally controlled by a positive
Most pigments are supplied as color blending step and re-sample exactly as
dispersions in alcohol, propylene before. (Note: sampling procedures and
glycol or water. sampling positions must be recorded).
Pigments reduce film strength. The content uniformity should not differ
Tablet coating pigments have a in both sampling sets of the two
significant effect on the film procedures.
properties. As pigments are added, a
reduction in flexibility and film
Revalidation
strength is experienced. Pigmented Infrequent re-blending should only
films exhibit a distinct loss of film occur in incidences of mechanical
strength from unpigmented films. breakdown.
Generally 20-30% additional Where the re-blending process
plasticizer (polyethylene glycol - becomes more frequent review and
PEG 600) is added when formulating revalidate the manufacturing process to
pigmented films. re-qualify the blending steps.
General Scale-up concepts COATING SUMMARY
The processing time for each
manufacturing step requires a ‘start and ¯ Hot water (80oC) dispersion is best
stop’ document entry. Where possible ¯ Allow to de-aerate before use
automatic recording control charts ¯ Continual stirring of coat maintained
should be in place for monitoring ¯ Filter (80 mesh) before use
processing times and critical process
temperatures. ¯ Quiescent period of 30-45 min.
Recording temperatures ¯Agitate coating solution continually
Temperature end-points for drying are during spraying
documented in the format of a range, ¯ Tablet friability - <0.5 - 1.0%
O O
i.e. ‘Inlet air 55 C (± 3 C) or heat bed ¯ Control core-bed load/weight
O O
to 42 C (± 2 C).’ It is not necessary to
¯ Avoid overloading or underloading
qualify both ends of such narrow
operating temperature limits (as there is ¯ Pre-warm core bed
no significant impact on the product or ¯ Jog bed until even temperature
process). distribution is obtained.
Re-mixing a step ¯ Set bed jogging cycle and jogging
Repeating a mixing or blending step is time to achieve correct bed temperature
acceptable, if the procedure has been ¯ Control inlet air temperature and RH
correctly qualified during the product ¯ Control inlet air volume
development phase.
¯ Control spraying rate (g/min)
To qualify a re-mixing or re-blending
¯ Control number of spray nozzles,
step manufacture the batch with the
height and angle.
initial mixing step. Sample the bulk at
the different sampling levels and ¯ Control nozzle distance form core
positions. bed.
Do not composite any samples for ¯ Insure smallest possible droplet size
content uniformity tests. ¯ Control outlet air temperature
With the same evaluation batch ¯ Insure a thermodynamic balance
immediately repeat the exact mixing or between heat in and water evaporation.
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
SC AL E UP P ROCEDURES
‘ the order is clear:- develop - scale-up - process qualification
- pivotal and validation lots ’
1. The process qualification batch is a ‘carbon copy’ of the pivotal lot? qYes qNo
2. The process qualification batch is manufactured under normal production qYes qNo
facilities ?
3. The process qualification batch documentation is similar or identical to the qYes qNo
pivotal lot documentation, which is identical to the commercial lots?.
4. Where two commercial batch sizes are manufactured, the equipment used qYes qNo
differs only in capacity or size?
5. Each batch size has a dedicated set of processing documentation? qYes qNo
6. All mixing/granulating times have a ‘start and stop’ entry? qYes qNo
7. All end-temperatures are documented with the recorded entries? qYes qNo
8. Speed, temperature and time control charts attached to manufacturing qYes qNo
instructions clearly identify batch, vessel, and processing step #?
9. Documentation records overall manufacturing and filling times ? qYes qNo
10. Special instructions exist to prevent product contamination during normal qYes qNo
processing breaks and temporary work stoppages?
11. Filling instructions document equipment cleaning and filling times? qYes qNo
12. Adequate controls exist to prevent over week-end/holiday manufacture? qYes qNo
13. Containers are air blown to reduce particulate matter and bioburden? qYes qNo
14. Line screen covers protect open containers from aerial particle settling? qYes qNo
Footnote : The Process Qualification Batch Documentation is the basis for the Pivotal manufacturing
documentation. Pivotal and commercial batch manufacturing instructions and procedures are in fact
identical in all respects with exceptions in equipment size changes. Both the process and the
manufacturing documentation under goes appropriate scale-up procedures.
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
S CA LE U P P ROCEDURES
Scale-up procedures
SOP
P-00-01-01Y2K Preparing a scale-up report for pivotal and validation batches.
P-00-01-01Y2K Time limitations between equipment cleaning and batch processing.
P-00-01-01Y2K Time limitations for completing overall granulation procedures.
P-00-01-01Y2K Maximum time between end of granulation and start of tabletting
operation.
Processing times:
The following pre-processing and processing controls have SOP documentation:
◊ maximum time period between equipment cleaning and batch processing
◊ the maximum period allowed between end of granulation and start of tabletting
operation.
◊ the maximum period allowed between end of tabletting and start of coating
operation.
4
[End of Document]
CLEANING
LIMITS
…‘Check the baby not the bath rinse water
Check the pot not the dish water…’
C
leaning validation is a focal contaminants from process equipment,
point of inspection for piping and hoses used.
regulatory agencies ∗ active material residues (limit)
throughout the world. For pre-
approval inspections conducted by ∗ excipient residues (non-soluble)
the FDA for any product ultimately ∗ Detergent and solvent residues
directed to the US market, many ∗ microorganisms (bioburden
companies have been denied reduction)
approval because of deficiencies in
their cleaning program or a lack of ∗ machinery lubricants
cleaning validation for their products. Solubility factors
This chapter reviews and highlights Non soluble active and insoluble
key regulatory expectations for a excipient residues require specific
satisfactory pre-approval inspection cleaning mechanisms to prevent cross
program. contamination or adulteration of the
next batch product. Maximum Residual
Validation Protocol limits are required to be established for
A cleaning validation program is an each active.
essential part of the generic product
Adulteration
development program. Zero adulteration for penicillin's has
Product approval has been denied been regulated. Steroids (e.g.
during pre-approval inspections hydrocortisone and estrogen) as well
(PAIs) conducted by the FDA due to as sulfa drugs require extremely low
the absence of appropriate scientific levels of active residues remaining in
rationale and documented the equipment after cleaning.
acceptance criteria for cleaning
program.
Written procedures
Written cleaning procedures (SOPs)
Residue types
are required detailing the cleaning
A well structured cleaning validation process for each piece of equipment
plan requires the removal after used in the manufacturing and filling
manufacture of the following principle process.
For cleaning detergent limits, a similar ratio employs current medical opinion
end point marker is used by along with appropriate safety factors
substituting toxic dose for therapeutic and allows a firm to defend its overall
dose. The cleaning detergent limit may cleaning program during regulatory
be defined or written as: review. Furthermore it allows the R&D
“the maximum contamination of detergent to development and coordinate and
residuals in the maximum daily dose for the standardize the scientific rationale for a
next product will not be more than 1/1000 cleaning validation program in all future
of its lowest toxic dose (LTD). ” product development.
ØC H E C K L I S T ×
SOP #P-HGD-03-01Y2K
1. Is there is a written cleaning procedure for every processing unit? qYes qNo
2. Is there a written cleaning procedure for all auxiliary equipment? qYes qNo
ØC H E C K L I S T ×
SOP #P-HGD-03-01Y2K
16.The 1/1000 of the lowest marketing dose (LMD) are the acceptable qYes qNo
active material residue limits ?
17.The 1/1000 of the LD50 are the acceptable detergent residue limits? qYes qNo
18. Wiping-solvents for cleaning swabs are specifically pre-determined qYes qNo
for each active material - subject to whether it's water soluble or insoluble?
19. Purified Water USP is the wiping solvent for most detergents? qYes qNo
23. Maximum time limits prior to cleaning have been established ? qYes qNo
24. Maximum time limits between equipment cleaning and sampling qYes qNo
have been established to avoid post-cleaning sampling errors ?
25. Maximum time limits between equipment cleaning and next batch qYes qNo
production have been established?
Use a combination of swabs and rinse waters tests
26. Equipment covers are used to protect exposed cleaned surfaces qYes qNo
during storage between batches (portable mixers etc.)?
27. Detergent concentration is standardized for soluble and non- qYes qNo
soluble active materials ?
28. Where applicable detergents and chlorine bleach are used qYes qNo
together?
29. Where applicable detergents and hydrogen peroxide are used qYes qNo
together?
30. Disposable clean cloths or filtered hot air is used for drying qYes qNo
equipment ?
31. Residual rinse waters are completely removed, after cleaning ? qYes qNo
STANDARD OPERATING
PROCEDURES
SOP #P-HGD-03-01Y2K Page 1 of 1.
CLEANING VALIDATION
…‘Time limits before and after cleaning must be laid down
in writing…’
4
[End of Document
CLEANING DEVELOPMENT
Cleaning Procedures
as the FDA See It
‘…These guidelines are intended for Bulk Pharmaceutical
Chemicals but almost every word applies to Drug Products…
V
alidation of cleaning procedures stated as follows "Equipment… shall
has generated considerable be maintained in a clean and orderly
discussion since agency manner…." A very similar section on
documents, including the Inspection equipment cleaning (211.67) was
Guide for Bulk Pharmaceutical included in the 1978 cGMP
Chemicals and the Biotechnology regulations.
Inspection Guide, have briefly Of course, the main rationale for
addressed this issue.
requiring clean equipment is to
CLEANING prevent contamination or adulteration
of drug products.
PROCEDURES Historically, FDA investigators have
must be validated looked for gross non-sanitation due to
inadequate cleaning and maintenance
These Agency documents clearly of equipment and/or poor dust control
establish the expectation that cleaning systems.
procedures (processes) be validated.
This guide is designed to establish
CLEANING
inspection consistency and uniformity PROCEDURES
must consistently
by discussing practices that have
been found acceptable (or
unacceptable). Simultaneously, one
must recognize that for cleaning meet the limits set
validation, as with validation of other Also, historically speaking, FDA was
processes, there may be more than more concerned about the
one way to validate a process. contamination of non-penicillin drug
In the end, the test of any validation products with penicillins or the cross-
process is whether scientific data contamination of drug products with
shows that the system consistently potent steroids or hormones.
does as expected and produces a A number of products have been
result that consistently meets recalled over the past decade due to
predetermined specifications.
address both scenarios and make it - FDA expects firms to conduct the
clear when a given procedure is to be validation studies in accordance with
followed. written cleaning protocols and to
Cleaning SOP document the results of studies.
- FDA expects a final validation
Water Soluble Residues report which is approved by
Cleaning SOP management and which states
whether or not the cleaning process is
Non-Soluble Residues valid.
Bulk pharmaceutical firms may decide The data should support a conclusion
to dedicate certain equipment for that residues have been reduced to an
certain chemical manufacturing "acceptable level."
process steps that produce tarry or IV. EVALUATION OF CLEANING
gummy residues that are difficult to VALIDATION
remove from the equipment. The first step is to focus on the
Fluid bed dryer bags are another objective of the validation process,
example of equipment that is difficult and we have seen that some
to clean and is often dedicated to a companies have failed to develop
specific product. Any residues from such objectives.
the cleaning process itself It is not unusual to see manufacturers
(detergents, solvents, etc.) also have use extensive sampling and testing
to be removed from the equipment. programs following the cleaning
Cleaning Limits process without ever really evaluating
the effectiveness of the steps used to
For 'Active Residues' clean the equipment.
Several questions need to be
& Limits addressed when evaluating the
For 'Cleaning Agents' cleaning process. For example:
Q. At what point does a piece of
What Do The FDA Expect equipment or system become clean?
- FDA expects firms to have written Q. Does it have to be scrubbed by
general procedures on how cleaning hand?
processes will be validated. Q. What is accomplished by hand
- FDA expects the general validation scrubbing rather than just a solvent
procedures to address who is wash?
responsible for performing and Q. How variable are manual cleaning
approving the validation study, the processes from batch to batch and
acceptance criteria, and when product to product?
revalidation will be required. The answers to these questions are
- FDA expects firms to prepare obviously important to the inspection
specific written validation protocols in and evaluation of the cleaning process
advance for the studies to be since one must determine the overall
performed on each manufacturing effectiveness of the process.
system or piece of equipment which Answers to these questions may also
should address such issues as identify steps that can be eliminated
sampling procedures, and analytical for more effective measures and result
methods to be used including the in resource savings for the company.
sensitivity of those methods.
Determine the number of cleaning Also check the written and validated
processes for each piece of cleaning process to determine if these
equipment. systems have been properly identified
Ideally, a piece of equipment or and validated.
system will have one process for
cleaning, however this will depend on LIST ALL
the products being produced and
whether the cleanup occurs between Complex Cleaning
batches of the same product (as in a processes and
large campaign) or between batches
of different products. Validate
Minor Cleaning In larger systems, such as those
employing long transfer lines or
of Same Product piping, check the flow charts and
Does not Require piping diagrams for the identification
of valves and written cleaning
Validation or Limits procedures.
W hen the cleaning process is used Piping and valves should be tagged
only between batches of the same and easily identifiable by the operator
product (or different lots of the same performing the cleaning function.
intermediate in a bulk process) the Sometimes, inadequately identified
firm need only meet a criteria of, valves, both on prints and physically,
"visibly clean" for the equipment. have led to incorrect cleaning
Such between batch cleaning practices.
processes do not require validation
('Minor Clean'). LIST Maximum
1. Equipment Design
Examine the design of equipment, Time Limits Allowable
particularly in those large systems that Between Process End
may employ semi-automatic or fully
automatic clean-in-place (CIP) and Start of Cleaning
systems since they represent
significant concern. Always check for the presence of an
For example, sanitary type piping often critical element in the
without ball valves should be used. documentation of the cleaning
processes; identifying and controlling
When such non-sanitary ball valves
the length of time between the end of
are used, as is common in the bulk
processing and each cleaning step.
drug industry, the cleaning process is
more difficult. This is especially important for
W hen such systems are identified, it topicals, suspensions, and bulk
is important that operators performing drug operations.
cleaning operations be aware of In such operations, the drying of
problems and have special training in residues will directly affect the
cleaning these systems and valves. efficiency of a cleaning process.
Determine whether the cleaning W hether or not CIP systems are used
operators have knowledge of these for cleaning of processing equipment,
systems and the level of training and microbiological aspects of equipment
experience in cleaning these systems. cleaning should be considered.
residues from the manufacturing and are satisfactory and can be readily
cleaning processes at very low levels. used.
If levels of contamination or residual Advantages of direct sampling are
are not detected, it does not mean that that areas hardest to clean and which
there is no residual contaminant are reasonably accessible can be
present after cleaning. It only means evaluated, leading to establishing a
that levels of contaminant greater than level of contamination or residue per
the sensitivity or detection limit of the given surface area.
analytical method are not present in Additionally, residues that are "dried
the sample. out" or are insoluble can be sampled
Firms should challenge the analytical by physical removal.
method in combination with the [b]. RINSE SAMPLES
sampling method(s) used to show that Two advantages of using rinse
contaminants can be recovered from
samples are that a larger surface area
the equipment surface and at what
may be sampled, and inaccessible
level, i.e. 50% recovery, 90%, etc.
systems or ones that cannot be
This is necessary before any routinely disassembled can be
conclusions can be made based on sampled and evaluated.
the sample results. A negative test A disadvantage of rinse samples is
may also be the result of poor that the residue or contaminant may
sampling technique (see below). not be soluble or may be physically
4. Sampling occluded in the equipment.
There are two general types of An analogy that can be used is the
sampling that have been found "dirty pot." In the evaluation of
acceptable. The most desirable is the cleaning of a dirty pot, particularly with
direct method of sampling the surface dried out residue, one does not look at
of the equipment. Another method is the rinse water to see that it is clean;
the use of rinse solutions. one looks at the pot.
Surface Cleaning Check the Baby
can be checked by not the
Surface Swabs Bath Water
or Check to see that a direct
This would be particularly true for the In establishing residual limits, it may
bulk drug substance manufacturer not be adequate to focus only on the
where reactors and centrifuges and principal reactant since other chemical
piping between such large equipment variations may be more difficult to
can be sampled only using rinse remove.
solution samples. There are circumstances where TLC
Any indirect test method must have screening, in addition to chemical
been shown to correlate with the analyses, may be needed. In a bulk
condition of the equipment. process, particularly for very potent
During validation, the firm should chemicals such as some steroids, the
document that testing the uncleaned issue of by-products needs to be
equipment gives a not acceptable considered if equipment is not
result for the indirect test. dedicated.
V. ESTABLISHMENT OF LIMITS The objective of the inspection is to
FDA does not intend to set ensure that the basis for any limits is
acceptance specifications or methods scientifically justifiable.
for determining whether a cleaning VI. OTHER ISSUES
process is validated. It is impractical a. Placebo Product
for FDA to do so due to the wide In order to evaluate and validate
variation in equipment and products cleaning processes some
used throughout the bulk and finished manufacturers have processed a
dosage form industries. placebo batch in the equipment under
The firm's rationale for the residue essentially the same operating
limits established should be logical parameters used for processing
based on the manufacturer's product.
knowledge of the materials involved A sample of the placebo batch is then
and be practical, achievable, and tested for residual contamination.
verifiable.
It is important to define the sensitivity
Testing a
of the analytical methods in order to Placebo Batch
set reasonable limits.
Some limits that have been may only be
mentioned by industry representatives
in the literature or in presentations
Supportive Data
include analytical detection levels However, we have documented
such as 10 PPM, biological activity several significant issues that need to
levels such as 1/1000 of the normal be addressed when using placebo
therapeutic dose, and organoleptic product to validate cleaning
levels such as no visible residue. processes.
One cannot assure that the
Check the manner in which limits are contaminate will be uniformly
established. Unlike finished distributed throughout the system.
pharmaceuticals where the chemical
identity of residuals are known (i.e., For example, if the discharge valve or
from actives, inactives, detergents) chute of a blender are contaminated,
bulk processes may have partial the contaminant would probably not
reactants and unwanted by-products be uniformly dispersed in the placebo;
which may never have been it would most likely be concentrated in
chemically identified. the initial discharge portion of the
batch.
Analytical
However validation of new generic
products in the pipeline may not have
an official assay method.
Validation
A totally new assay method possibly
based on a Pharmacopeial Forum
methodology is adapted in-house for
Analytical Validation - a working assay as well as for stability indicating
validation protocol for HPLC system. testing methodology.
Analytical Aspects I
Validation of analytical methodology
for an HPLC system.
1. PURPOSE
The purpose of this Standard Analytical Procedure is to demonstrate the procedure
required to validate in-house HPLC analytical methods and to show that the methods
are stability-indicating. Methods based on the USP but modified for stability
indicating test purposes require full in-house validation.
This procedure ensures that the Product Development Process and Process
Qualification Batch analysis is based on a foundation of Good Laboratory Practice
using validated test procedures.
2. RESPONSIBILITY
The Head of Analytical Development in coordination with the managers of QC and
Regulatory Affairs at the proposed manufacturing site.
3. FREQUENCY
For each non-compendial analytical method intended for ANDA (or OTC ANDA)
manufactured products.
For Stability-Indicating Assays and limit testing of impurities that may be based on
compendial methods. Each Product strength will follow the full method validation
procedure.
4. PROCEDURE
[a]. Method Validation
Non-compendial methods validation will follow the USP direction for parameters
needed for the validation of test methods.
Typical parameters for validating assays and other non-compendial analytical
methods designed for providing quantitative results shall include :
• Accuracy
• Recovery
• Precision ( System reproducibility, Method reproducibility )
• Specificity
• Linearity
• Range
• Ruggedness (different analysts / days /different equipment models / columns)
Standard Solutions are stored at controlled temperatures and light conditions as per
labeling.
The values depict the specificity of the method for resolution between the main peak
and impurity peak. (values shown for demonstrations purposes).
Peak Purity
The photo diode-array is used for the evaluation of the stability indicating nature of
the assay method number SI-1000 for [000]mg and [000]mg tablets using a Waters
996™ Unit, controlled by the chromatography manager Millennium 2010™.
Peak purity and match results are reported as:
Purity Threshold is the sum of Noise Angle and Solvent Angle. It is the limit of
detection of shape differences between two spectra.
Match Angle is a comparison of the spectrum at the peak apex against a library
spectrum.
Match Threshold is the sum of the Match Noise Angle and Match Solvent Angle.
Analytical Aspects II
Validation of analytical methodology
for an HPLC system.
Precision - Table 1.
System Repeatability
[Also called intra-assay precision]
SYSTEM REPEATABILITY
SAMPLE No. PEAK AREAS
1
2 Repeatability shows
3 precision
4 under the same operating
5 conditions over a short
6 interval of time - same day
7 same morning
8
9
10
Average Peak Area
Standard Deviation =
Relative Standard Dev. =
= 0.5 - 1.0
METHOD REPRODUCIBILITY
SAMPLE No ASSAY %
Batch No:
1
Method Reproducibility
2 monitors the sample-to-
3 sampling variation of the
4 same drug product by
5 evaluating the full
6 analytical method over
7 and over again.
8 (same operator - same
9 equipment)
10
Average Assay % =
Standard Deviation =
Relative Standard = 1.5 - 3.0
Deviation.
[4] Accuracy
The Accuracy of an analytical procedure expresses the closeness of agreement
between the true value and the value found.
Ten replicate (single) injections of the standard solution at the nominal concentration
of x mg/100 mL as described in the Analytical Method / Ed is made and the percent
deviation from the true values as determined from the linear regression line is
calculated.
♦ The Results (Peak areas and % accuracy) are tabulated.
♦ The Mean, SD and C.of.V are shown in the tabulations
ACCURACY
1
2
3
4
5
6
7
8
9
10
Mean (% Accuracy) =
Standard Deviation =
% Coef. of Variation =
RECOVERY - EXTRACTION
TIME IN
MINUTES % ASSAY
Batch No:
1.5 T
R E C O V E R Y
Standard solution mg/100mL Peak Area =
50
75
100
125
150
Mean (% Recovery) =
Standard Deviation =
% Coef. of Variation =
Display values
The Linear Regression value, Slope and Y-Intercept are shown in the GRAPH. The
placebo chromatogram (vehicle only) is shown to highlight the absence of Additional
Peaks
Linear Regression =
Y-Intercept =
Slope =
The results are shown graphically (peak area Vs range conc. (mg/100 mL).
GRAPH OF LINEARITY
120000
P
e 100000
a
80000
k
60000
A
40000
r
e 20000
a
0
0 25 50 75 100 125 150
Conc. mg/100mL
Robustness is defined by both the USP and the ICH Tripartite guidelines as "a
measure of its capacity to remain unaffected by small but deliberate variations in
method parameters and provides an indication of its reliability during normal use "
Robustness is defined both in the USP and ICH, but is not required.
Robustness variations.
Deliberate variations according to the following table were made to the critical
parameters of the method such as column, flow rate and concentration of [organic
acid] in the mobile phase. Using the System Suitability solution and LOQ (also QL)
solution as the Test Solutions the performance of the method was evaluated.
ED. N0: 02 Effective Date:
Replaces Ed 01.
APPROVED:
Ed. Status: MM/DD/2000
Operational
Department R&D QC QA
Handbook of Pharmaceutical Sect: 13.16
13.16 Generic Development
ORAL TABLETS ANALYTICAL CHAPTER 13
RUGGEDNESS
ANALYST % ANALYST %
No 1 ASSAY No 2 ASSAY
Column I Column 2
1
2
3
4
5
6
7
8
9
10
Mean (% Accuracy) =
Standard Deviation =
% Coef of Variation =
STABILITY OF
THE STANDARD SOLUTION
mg/100mL mg/100mL
Initial Analysis Repeat Analysis
(Date) 2nd (Date)
1 injection 1 injection
2 injection 2 injection
3 injection 3 injection
4 injection 4 injection
5 injection 5 injection
6 injection 6 injection
7 injection 7 injection
8 injection 8 injection
9 injection 9 injection
10 injection 10 injection
Mean =
Standard Deviation =
Relative Standard Dev. = NMT 2.0 %
Representative chromatogram
Drug Product
Revalidation
An entire validation procedure should Validation Checklist
be repeated again for new approved 1. Are all in-house methods validated?
sources of the active material as the 2. Is the dissolution method for
new active may well display a different development, release and stability
impurity profile and an altered validated.
degradation pattern under stressed 3. Are the different validation editions
conditions. comparable with the previous
edition?
All of the above validation parameters 4. Was the stability assay and
are mentioned in the SUPAC (3) guide dissolution method validated at the
for analytical methods, developed for time of process optimization
bioequivalence blood sample testing: 5. Was the stability assay and
i.e. dissolution method validated at the
♦ Accuracy time of process qualification
6. Is the validated stability assay,
♦ Specificity
impurity profile and dissolution
♦ Recovery method used in the pivotal essential
♦ Precision (interday & intraday) similar to the commercial validation
♦ Linearity (of standard curves) lots so that data is comparable.
♦ Sensitivity 7. When a new edition is not
♦ Stability (Storage & handling) comparable to the previous edition -
is a new method number allocated?
8. Has the lab an historical track record
Bioequivalence studies of all assay dissolution etc. methods
Linearity and range at the extreme
used from early development to
lower ends of the active analyte or
commercial validation?
metabolite(s) in sera are important
parameters in assaying blood serum References:
concentrations.
1. "Validation of compendial methods" USP 23
Range studies MUST be linear at the <1225> USPC Rockville Maryland USA
lower and upper limits as found in the 1994.
serum samples or those used for 2. USP/NF XXIII USPC Rockville Maryland
USA 1994.
calibration curves, a point sometimes 3. Scale up and Post approval Changes
frequently overlooked by analytical Manufacturing and Controls In vitro
method developers in clinical Dissolution and In Vivo Bioequivalence
environments when dealing with very, Documentation CEDER 1995 (SUPAC)
4. International Conference on Harmonization
very dilute concentrations. "Guidelines on validation of Analytical
Procedures: Definitions and Terminology;
All analytical validation attributes Federal Register (March 1, 1995.)
require to be taken into account, 5. ASTM Standard Guide For Conducting
including ruggedness. No robustness Ruggedness Tests E1169 American Society
tests are officially required for USP and for testing Materials Philadelphia 1989.
6. G. Kateman and L. Buydens, The
ICH, however prudent analytical Ruggedness Test Quality Control in the
method developers should build in a Analytical chemistry John Wiley and Sons
robust test, whether official or not. NY 2nd Edition 1993, pp118 125.
OUT-OF-SPECIFICATION
TEST RESULTS
‘…Investigate all Out-of-Specification Results routinely and immediately,
the findings may well be in the firms commercial interests...’
Out Of Specification
P
racticing the correct do’s and don’t
procedures in out-of-specification
decisions making is a central
(OOS) Retesting and
requirement for an analytical laboratory Product failures based
personnel. The FDA 1998 draft OOS on the 1993 Judgment
guidelines applies to all drug product Drug developers and manufacturers
manufacturers and may be a useful tool should be thoroughly conversant with the
for the day-to-day administration and FDA's September 1998 draft 'Out-of-
decision making in a research or routine Specification' guidelines and current
quality control analytical laboratory (see thinking which may provide an appropriate
OOS checklist). base for decisions in routine QC and
Judge Alfred M Wolin’s original 1993 production OOS investigations.
interpretation of Good Manufacturing An Out of Specification
Practice (GMP) issues embedded in the
landmark US court ruling five and half (OOS) Result
years ago is the underlying basis of the may or may not
FDA's September 1998 DRAFT Out-of-
Specification guidelines for the generic be a product failure
and innovative pharmaceutical industry. Out-of-Specification results are not
Notwithstanding that the court ruled in necessary product failures, a term FDA
April 1993 on important OOS laboratory investigators commonly use in PAI and
practices, FDA EIRs and 483 Inspection GMP inspections. Handling out-of-
observations, still cite non-compliance specification lab data has become a
practice with respect to product retesting primary GMP inspection focal point.
and Out-of-Specification (OOS) test The failure for some firms to conduct an
results in both Generic and Innovative adequate data investigation has become a
drug firms. frequent citation in PAIs and routine GMP
The interpretation and application of the Agency Inspections.
Good Manufacturing Practices (GMP) Act Where is my data
have set practical ‘hands-on’ guidelines invalidating
that support the industry rules for handling
out-of-specification test results, retesting
an Out-of-Specification
procedures and product failure (OOS) Result?
investigations. Out-of-Specification results
These rules are now incorporated in the must not become ‘un-investigated
draft 'guidance for industry investigating failures’. The maxim “we investigated but
out-of-specification test results for didn’t write it down” is often heard and
pharmaceutical industry'. carries little weight with an agency official.
What the PAI investigator really hears is, and has no scientific merit or statistical
“we didn’t bother to investigate and don’t value).
have any SOP requiring us to do so”. For practical purposes this invalid result
Such an attitude is in violation of the new was technically never done and should
draft guidelines and the firm needs to not be used for any calculation or
address its investigation procedures statistical evaluation. However the
impacting on product retesting and analyst error must be thoroughly
evaluating drug product approvals. documented and properly invalidated -
Out-of-Specification Test Results (OOS) may with written reasons, supervisor and
be:- Reversible due to a:- analyst signatures and date of
invalidation process.
Ü genuine laboratory error
Ü sampling error
Investigate according
Or non- reversible due to a :- to a set procedure
Ü manufacturing processing error and then retest if the
Ü manufacturing operator error error is conclusive?
Genuine Laboratory Error. Inconclusive errors
Laboratory errors can occur when Where an error is investigated and the
analysts make analytical mistakes. All result of the investigation is inconclusive
laboratory technicians and analysts may the following rules apply to the retest
mistakes at times in their career. There is procedure.
no such concept as an error-free Inconclusive errors retest
laboratory analyst or that all standard
solutions are always correct and that data An inconclusive error is an OOS where
is never miscalculated. Samples may be the 'supervisor-analyst investigation' did
incorrectly prepared, diluted, injected or not draw a firm conclusion and the reason
stored at inappropriate environmental for the error was not clearly identified.
temperatures or quite simply that What to do: Answer - Retest with new
containers where not properly closed or aliquot (replicates, if required) from the
even placed in the correct designated same sample, if the sampling procedure
sampling container. was proven OK by investigation. If the
A suspected laboratory error must be sampling procedure was found to be in
investigated and if found genuine can error, then re-sample the target material
immediately be invalidated and the result and perform a new duplicate analysis.
simply filed and disregarded completely.
The investigation or ‘failure investigation’ For process failures
should where ever possible identify the confirmed by laboratory
cause of the OOS and its impact. testing - means the
Retest procedures product has failed?
must follow DECISION TREE
pre-written rules An overview of Out-of-Specification
Once the nature of the OOS has been Results procedures is provided by the
decision tree.
identified - as an laboratory error - a
Re-sampling the material for a new
repeat test must be performed and the representative sample should take place only
initial test totally ignored. (Remember this when the original procedure was found to be
initial test result is invalid - it was an error clearly non-representative of the whole.
Do’s
& Don’ts
The ‘Retesting Rules’: Retest procedures
Do insure that the firm has a written
must follow
Retest SOP. pre-written rules.
Do insure the retest SOP indicates when The Don’ts
testing stops and the product is evaluated. Don’t - use the outlier test for a
Do retest only after; dissolution test or profile.
Don’t - use the outlier test for an
⇒ reporting,
Assay or a Content Uniformity test.
⇒ classifying and Don’t - use the outlier test frequently
⇒ investigating jointly. to reject non-chemical test results.
Don’t - use the outlier test outside
Do retest on the same sample
the USP specifications prescribed.
collected.
Don’t - retest before an investigation
Do feel free to prepare a second aliquot is completed and closed.
from the same sample as the first.
Don’t - retest before reporting to
Do use a larger portion of the first your supervisor and conducting an
sample, if required. informal investigation - together.
Do know when you can and can’t use Don’t - ignore ever the rules for
governing a single or multiple OOS result.
the outlier test.
Don’t - re-sample unless the sample
Do re-sample if the original sampling procedure was proved to be faulty.
procedure is proven by investigation, as
not representative. Don’t - re-sample without your
supervisor permission obtained only after
Do re-sample if the original sample was an informal investigation.
improperly prepared (clearly shown after
the required investigation). Averaging passing and
OOS results together
Do investigate and then close the
investigation within 30 days after the
is not GMP
OOS. Don’t - Average good and bad
results - as they simply hide the true test
Do keep a ‘Failure Report” or an values. 3
‘Investigation Report’ log.
ØC H E C K L I S T ×
CL # P-HGD-03-1Y2K
OUT-of-SPECIFICATION RESULTS
‘…Averaging passing and OOS Test Results together
is not permitted as it conceals the full analytical picture…’
ØC H E C K L I S T ×
CL # P-HGD-03-1Y2K
OUT-of-SPECIFICATION RESULTS
‘…failure investigations are conducted to determine
what caused the unexpected OOS result…’
ØC H E C K L I S T ×
CL # P-HGD-03-1Y2K
OUT-of-SPECIFICATION RESULTS
OUT-OF-SPECIFICATION
TEST RESULTS
‘…An Out-of-Specification Test Result is not necessary
a product failure - and needs to be qualified …
and should involve all other [2]. Testing a specimen from the
departments that could be implicated, collection of a new sample from the
including manufacturing, process batch
development, maintenance, and [3]. Re-sampling testing data
engineering. Other potential problems [4]. Using outlier testing.
should be identified and investigated.
1. RETESTING
QC Unit Investigates:
Part of the investigation may involve
Production and Laboratory retesting of a portion of the original
sample. The sample used for the
Systems & Documentation retesting should be taken from the
same homogeneous material that was
The records and documentation of the originally collected from the lot, tested,
manufacturing process should be fully
and yielded the OOS results.
investigated to determine the possible
cause of the OOS results. For a liquid, it may be from the original
[A]. General Investigational Principles unit liquid product or composite of the
liquid product; for a solid it may be an
A failure investigation should consist of additional weighing from the same
a timely, thorough, and well- sample composite that had been
documented review. prepared by the analyst.
The written record should reflect that
the following general steps have been Situations where retesting is indicated
taken. [ I - T R A C ] include investigating testing instrument
[1]. The reason for the Investigation has malfunctions or to identify a possible
been clearly identified. sample handling integrity problem, for
[2]. The overall manufacturing process example, a suspected dilution error.
sequences that may have caused the Generally, retesting is neither specified
problem should be summarized. nor prohibited by approved applications
[3]. Results of the documentation review or by the compendia.
should be provided with the assignment of
actual or probable cause.
Investigation Retesting:
[4]. A review should be made to determine First Performed on
if the problem has occurred previously.
[5]. Corrective actions taken should be the original sample
described. by a Second Analyst
The general review should include a Decisions to retest should be based on
list of other batches and products the objectives of the testing and sound
possibly affected and any required scientific judgement. Retesting should
corrective actions taken including any be performed by an analyst other than
comments and signatures of the one who performed the original
appropriate production and quality test.
control personnel regarding any
material that may have been
The CGMP regulations require the
establishment of specifications,
reprocessed after additional testing.
standards, sampling plans, test
[B]. Laboratory Phase of an Investigation
procedures, and other laboratory
A number of practices are used during
control mechanisms (§ 211.160).
the laboratory phase of an
investigation. These include: The establishment of such control
[1]. Retesting a portion of the original mechanisms for examination of
sample additional specimens for commercial or
regulatory compliance testing must be
[A]. Interpretation of Investigation Results [2] does not confirm the OOS result —
An OOS result does not necessarily the OOS result should be retained in
mean the subject batch fails and must the record and given full consideration
be rejected. in the batch or lot disposition decision.
OUTLIERS
MAYBE:
OUTLIERS OUTLIERS
CAN BE:
TEST SAMPLE
DEVIATIONS VARIABILITY
Don't assume an
way out result must be
a testing error UNIFORMITY
DISSOLUTION
OF
TESTING
CONTENT
(COURTESY: IJGD
Vol. 02 #07 (1998)
international Journal of Generic Drugs
YES
B A
PRODUCTION Investigate LABORATORY
OOS NO NO
Clear laboratory
Retest
error
YES
YES
SPECIFICATION
FAILURE Same Sample
Different Analyst Invalidate
(Evaluate Batch result & retest
for Rejection) (DO NOT RETAIN RESULT)
IF
OOS NO
YES
The
RE-SAMPLE
New representative
specimen
IF
YES OOS NO
Don't Apply Outliers To:
Content Uniformity
Dissolution test
Composite Assays
ASSAY VALIDATION
Ruggedness
and
Robustness
and appraised individually. A simple Even more dramatic, for eleven variables
experimental design can evaluate both (11 x 12) a minimum of 132 one-factor-at-a-
ruggedness and robustness, as separate time data points would be required, but via
distinguishable entities - together. matrix testing using an twelve-run design,
only 12 HPLC assays are needed to produce
Table 1 Comparison Table. the equivalent of 132 individual one-factor-
Attribute. Ruggedness. Robustness. at-time assays.
USP Validation þ ý
Few, if any HPLC assay analytical methods
Requirement could have more than 12-15 significant
ICH Validation environmental or method variations.
ý ý
Requirement Table 1.
Internal change ý þ An Eight Run Design Template
External / Internal ASSAY
External change þ - Test Changes / Variations TEST
Method - þ No A B C D E F G RESULT
Variations
Environmental þ - 1 + + + − + − − 99.3
Variations 2 − + + + − + − 101.5
3 − − + + + − + 100.4
Designing Analytical Experiments
4 + − − + + + − 97.9
Such a designed experiment can
demonstrate that methodology and 5 − + − − + + + 98.5
environmental factors may or may not 6 + + − − + + 99.0
influence the test results. It is hoped that the 7 + + + − − + 97.9
analytical method is both rugged and robust,
however a well designed experiment may 8 − − − − − − − 100.9
identify test conditions or specification limits The (+) or (-) signs are used as variables in the 8 run
design. Assign (-) to Analyst I ; Day I; Column I and
that need to be closely controlled and (+) to Analyst II; Day II; Column II and so on
tightened or even test parameters that need A to G are chosen as the external variations
further investigation and optimization. (ruggedness) anticipated to arise during use in the
Development Lab.
Advantages - Designing the Experiment Table 2. 8 RUN DESIGN
A designed experiment is a simple matrix
design. The Plackett-Burman designs are Template for Ranked Effect and Means
most applicable to technical transfer of a External & Internal Ranked M
validated analytical methods from changes/variations Effects values
development to quality control centres. For
A - Analyst I & II 1.8 -1.35
the most cost-effective design, the attributes
of both R&D and QC laboratories should be F - Analyst III & IV -0.76
incorporated into development validation G - Reagents I & II -0.35
protocol of the assay method. E - Week I & II 0
The advantages of these designs are quite B - Week III & IV +0.35
simple - the number of tests required is
C - Column I & II +0.76
simply dramatically reduced. 56 assays (i.e.
7 x 8) are needed to evaluate seven internal D - HPLC No I & No II 0 +1.35
and/or external variables, these can be The M values are obtained from statistical design
reduced to eight quick assays using an eight- tables. The ranked Effects are calculated by simple
addition of assay test results and then dividing by
run Plackett-Burman design.
half the number of runs (i.e. 4 in a 8 run design).
Method Procedure.
1. Choose the number of variables required Process Qualification Stage.
and select a run design template. The evaluation of ruggedness and
robustness should be finalised at the end
2. Assigning the minus (-) or plus (+) values: of the development phase - around the
These are arbitrary designations. As a time of the process qualification lot
standard rule assign a 'minus' (-) to I or a
manufacture. The
lower limit and a 'plus' (+) to II or a upper
ruggedness/robustness evaluation should
limit. Evaluate a range limit by assign (-)
value for lower and (+) value for higher (i.e. be developed with the commercial
Flow rate 1.2 mL/min assign (-) and 1.8 laboratory equipment in mind. It should
mL/min assign (+)). Likewise Day I assign (- show the reliability of an analysis with
) and Day II assign (+) and so on… respect to deliberate variations in the
method parameters.
3. Perform the HPLC assays in a random
order. Ruggedness/robustness determinations
are essential when transferring analytical
4. Tabulate the assay results in the template.
methods from the development
5. Calculate the Effects (Figures 1 and 2). laboratory to the commercial plant
6. Rank the Effects from smallest to largest. quality control laboratory. There may
usually be a difference in columns or
7. Plot the Effects against the M values.
HPLC machine models used.
8. Evaluate the plot. A consequence of ruggedness /
Conclusion. robustness evaluation is that a series of
The results from the plot form a near system suitability parameters are
straight line. It can be concluded that the established to ensure that the validity of
analytical method is (a) rugged for the the analytical procedure is maintained
external factors over the tested range and whenever used.
(b) robust for the internal factors over the
tested range in the 12 run design.
References:
Figure 3.
1. "Validation of compendial methods" USP 23
A Normal Probability Plot of Effects <1225> USPC Rockville Maryland USA 1994.
2. International Conference on Harmonization
+1.5 "Guidelines on validation of Analytical
* Procedures: Definitions and Terminology…;
M Federal Register (March 1, 1995.)
* 3. "Validation of compendial methods" USP 23
V <1225> USPC Rockville Maryland USA 1994.
A * 4. USP/NF XXIII USPC Rockville Maryland USA
L 1994.
U * 5. Scale up and Post approval Changes
E Manufacturing and Controls In vitro Dissolution
S * and In Vivo Bioequivalence Documentation
CEDER 1995 (SUPAC)
* 6. ASTM Standard Guide For Conducting
Ruggedness Tests E1169 American Society for
* testing Materials Philadelphia 1989.
-1.5 7. Kateman and L. Buydens, The Ruggedness Test
Quality Control in the Analytical chemistry John
-8 Ranked Effects +7
Wiley and Sons NY 2nd Edition 1993, pp118
125.
ANDAs Impurities in
Drug Substances.
IAGIM Scientific Committee GUIDANCE FOR INDUSTRY
Block JD; Holmann E ; West P
INTRODUCTION
TABLE OF CONTENTS - 11/1999
Evaluate & Compare
I. INTRODUCTION
Innovator Drug
II. CLASSIFICATION OF IMPURITIES
III. RATIONALE FOR THE REPORTING AND
CONTROL OF IMPURITIES
Impurities
A. Organic Impurities
B. Inorganic Impurities
CHEMISTRY ASPECTS
C. Residual Solvents including classification and
IV. ANALYTICAL PROCEDURES identification of impurities, generating
V. REPORTING IMPURITY CONTENT OF reports, setting specifications, and a
BATCHES brief discussion of analytical
VI. ACCEPTANCE CRITERIA FOR IMPURITIES procedures; and
VII. QUALIFICATION OF IMPURITIES SAFETY ASPECTS,
VIII. NEW IMPURITIES including comparative studies and
ATTACHMENT I — Impurities Decision Tree (Generic genotoxicity testing. Specific guidance is
Drug Substance)
provided for:
ATTACHMENT II — ICH Decision Tree for Safety
Studies l Qualifying impurities found in a drug
ATTACHMENT III — Glossary substance used in an ANDA by a
comparison with impurities found in the
ANDAs: Impurities in Drug related U.S. Pharmacopeia (USP)
Substances. [Nov 1999] monograph, scientific literature, or
INTRODUCTION innovator material;
Residual solvents
(e.g., equivalent detector response).
These assumptions should be
have their own discussed in the DMF submission or
abbreviated application.
guidelines V. REPORTING IMPURITY CONTENT
IV. ANALYTICAL PROCEDURES OF BATCHES
The DMF or abbreviated application Analytical results should be provided
(ANDA) should include documented for all batches of the drug substance
evidence that the analytical procedures used for stability testing, as well as for
are validated and suitable for the batches representative of the proposed
detection and quantitation of impurities. commercial process.
impurity is a impurity
specified.
Erratic
specified in the batch-to-batch
drug monograph impurity levels
(identified or unidentified) may indicated
Although normal manufacturing incomplete validation
variations are expected, significant
variation in batch-to-batch impurity The DMF holder or the ANDA applicant
levels could indicate that the should provide a rationale for selecting
manufacturing process of the drug impurity acceptance criteria based on
substance is not adequately controlled safety considerations.
and validated. The level of any impurity present in a
In summary, the drug substance drug substance that is in compliance
acceptance criteria should include, with a USP specification or has been
where applicable, acceptance criteria adequately evaluated in comparative or
for: in vitro genotoxicity studies or has been
would then fall outside the purview of The impurity is qualified if it is found at
section 505(j) of the Federal Food, similar levels (no more than twofold
Drug, and Cosmetic Act (the Act). higher, but not to exceed 1.0% for most
Additional clarification regarding the drug substances). Twofold higher
levels in the Impurities Decision Tree for criteria are justified for several reasons.
Unidentified
generic drug substances is provided
below:
The Levels impurities present
Ø First level (L1): Is the impurity in
question "above threshold"? (See the in both innovator &
generic are deemed
threshold table in section VII.)
This level is identical to the
corresponding level in the ICH Decision acceptable
Tree for Safety Studies (Attachment II).
Ø Second Level (L2): For example, the innovators' impurity
acceptance criteria are set higher than
Is the "structure elucidated"? levels observed in drug substances, and
This refers to structural identification or the safety studies that qualified the
characterization exactly as in the ICH innovators' drug substances are carried
Decision Tree for Safety Studies. out at significantly higher levels than the
However, in those rare cases where it is specifications agreed to under FDA's
not possible to identify the impurity by pharmacology and toxicology
structure, the efforts made should be evaluations.
satisfactorily documented. In certain dosage forms where
Once the impurity has been structurally sensitivity or toxicity concerns arise, the
identified, one could go to level L3. impurity levels should be no higher than
Ø Third Level (L3a): Compliance with a the innovator's level for toxic impurities.
USP acceptance criterion for a known In generic drugs, an unidentified
individual impurity (e.g., see impurity impurity may still be considered
listed in the Clidinium Bromide USP qualified in cases where the impurity is
monograph). observed at similar levels in the
innovator's product via a comparative
Evaluate 3 or more study.
innovators lots to Unidentified
establish ANDA's impurities present in
impurity baseline
the innovator drug
Third Level (L3b): A comparison of the
impurity profile of the generic drug are qualified
substance with the process impurities Third Level (L3c): This level looks at an
profile on an average of three or more
impurity at a "higher level, or a different
different lots of the innovator's drug
new impurity."
product is recommended.
New means one that was not previously
This comparative study should be seen in the bulk drug substance.
performed using appropriate The level of the new impurity may be
discriminating analytical tests such as
qualified from the scientific literature if it
HPLC or Capillary Electrophoresis.
is substantiated that this impurity is an
Yes
Is the impurity
No No observed in the No
Decrease below
L2 Structure elucidated? innovator’s drug
threshold
product and at a
similar level?
Yes
No Yes
No
No
Yes
No
Structure elucidated?
Yes
No
No No Yes
Adverse Effects
Yes No
a If considered desirable, a minimum screen for genotoxic potential should be conducted. A study to detect point
mutations and one to detect chromosomal aberrations, both in vitro, are seen as an acceptable minimum screen.
b For NDAs, if general toxicity studies are desirable, study(ies) should be designed to allow comparison of
unqualified to qualified material. The study duration should be based on available relevant information and
performed in the species most likely to maximize the potential to detect the toxicity of an impurity. In general, a
minimum duration of 14 days and a maximum duration of 90 days will be acceptable.
ORAL TABLETS ANALYTICAL DEVELOPMENT CHAPTER 13
Post-approval Changes in
Analytical Testing Laboratory Sites
'PAC-ALTS'
ØC H E C K L I S T ×
CL # P-HGD-01-01Y2K Page 1 of 1
1. This change procedure applies to approved drug products only. qYes qNo
2. Applies to all analytical tests at any stage of drug product's life-cycle qYes qNo
- from raw material analysis to post approval stability.
3. A copy of the laboratory's Quality Assurance Profile (QAP) from the qYes qNo
FDA's Freedom of Information (FOI) Office.
4. The chosen laboratory passed a satisfactory cGMP inspection within qYes qNo
the last two years (See attached SOP for IQOQ qualification).
5. No objectionable conditions or practices were found during the last qYes qNo
inspection.
6. Where objectionable conditions were found, adequate corrective qYes qNo
action has been taken to satisfy the FDA's initial objections.
7. Analytical testing Laboratory (ATL) must prove test can be fully qYes qNo
performed with accuracy and precision.
8. ATL must prove its testing capability for each and every test. qYes qNo
9. ATL must have a recorded history of performing the specific test(s) qYes qNo
(i.e. a written track record.)
10. ATL must have appropriate SOPs for the specific test(s) and qYes qNo
procedures.
11. ATL must have suitable calibrated equipment for the test procedures qYes qNo
12. ATL must have suitable information to support its capability to qYes qNo
perform the intended analytical testing procedures.
13. ATL must have trained personnel to perform the test procedures qYes qNo
(availability of updated training records, specific to the required
analysis)
14. ATL overall capabilities must be available for agency inspection. qYes qNo
15. FDA will be informed of the change being effected, via the Annual qYes qNo
Report or a future Supplement (whichever is submitted sooner.)
16. Where a supplement is filed, it is clearly identified in the heading and qYes qNo
text as being filed under PAC-ATLS rules - and,
17. A written statement is included why a PAC-ATLS CBE supplement qYes qNo
is appropriate (i.e., the four circumstances listed above, all exist.)
ATTACHMENT ONE
(This attachment consists of 12 pages)
EQUIPMENT IDENTIFICATION
Description
Manufacturer
Equipment ID No:
CONTENTS
PROCEDURE COMPLETED
n Approval Document Completed q
¯ Conclusion Completed q
Approval Document
According to the data collected as a result of the protocol, this equipment as defined
under equipment identification has been properly installed and operationally
qualified in accordance with the attached Standard Operational procedures in
practice.
Option 1
n Installation Qualification has been carried out prior to the use Yes q NO q
of the equipment.
Option 2
n Installation Qualification has been carried AFTER to the use of Yes q NO q
the equipment.
Option 3
n The equipment has been fully qualified and calibrated Yes q NO q
Option 4
n The equipment is now fully qualified and is suitable for use Yes q NO q
AUTHORIZATION
On completion of Option 3 the equipment is dually authorized by signing the
authorization and approval footer at the end of each page.
Installation Document
According to the manufacturer's specification this equipment has been properly
installed and is in compliance with the attached Standard Operational procedure.
INSTALLATION
UNIT SPECIFICATION APPROVAL STATUS
EQUIPMENT CHANGES
CHANGES TO THE DOCUMENT Reviewer's Year
LABORATORY EQUIPMENT REFERENCE Signature
1999
Major or fundamental
changes may require a
complete re-qualification
(if equipment has a 2000
different operating
principle)
2001
After 5 years have elapsed
examine the documents and
equipment in order to evaluated
if a full re-qualification is 2002
necessary
2003
Operational Test
OPERATIONAL TEST
OBJECTIVE
To test the operation of the equipment under normal operating conditions.
TEST RESULTS
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
Approved q Failed q
COMPLETED BY DATE:
n Display summary of test results certifying that for each laboratory model or pilot
plant unit - indicating that equipment has been:-
- Qualified or re-qualified (IQOQ)
- Cleaning program results are within specification
- Routine equipment maintenance program functional
- Calibration program operational and effective
- SOP review has been undertaken and signed-off according at the time of review.
CONCLUSIONS
n This Installation & Qualification Program applies to: Laboratory / pilot plant
instrumentation and equipment.
The equipment has been Installation & each model type operationally qualified. The
following real time systems have been put in place.
[1] An Audit and Check Program has been installed to ensure that appropriate
IQOQ (or re-qualification) program has been executed according to the current
IQOQ SOP.
[2] An Audit and Check Program has been installed to ensure that an appropriate
MAINTENANCE PROGRAM has been put in place for relevant equipment.
[3] An Audit and Check Program has been installed to ensure that an appropriate
CALIBRATION program has been put in place for relevant equipment.
[4] An Audit and Check Program has been installed to ensure that appropriate
PERFORMANCE VERIFICATION program has been put in place for relevant
equipment.
COMPLETED BY DATE:
CHECKED BY DATE:
Installation
Operational Test
Tests Performed
In-house test methods
performed according to protocol
Kept on file in metrology /
engineering department as
prescribed in the SOP.
COMPLETED BY DATE:
Installation
Describe the installation
level relevant to each
responsible person
(What did each contracting
installer actually do)
Operational Test
Tests Performed
Third party test methods
to be kept on file in
metrology / engineering
department / or signed
summaries.
An appendix to this report stating that all non-employees as listed above are
competent to perform the installation or operational qualification work pertaining to
this equipment.
COMPLETED BY DATE:
APPENDICES
LIST OF APPENDICES TO THIS REPORT ATTACHED HERE
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
List the number of
11.
appendices and attachments
12. affixed to this report at each
13. time period.
14.
15.
3
[End of Document]
Process
QUALIFICATION
‘…The process qualification batch is a simulation of the forthcoming pivotal
l o t proving in-house, that the process really works…‘
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
PROCESS QUALIFICATION
‘…well developed process qualification batch with good in-process
controls
and excellent documentation will ensure a failure-free pivotal lot…’
1. The Process Qualification batch is equal to the 70% or more of the pivotal qYes qNo
batch or the smallest commercial batch size that will be validated?
2. The active material source has been verified as an ‘Approved Supplier’ ? qYes qNo
3. All non actives are routine production excipients or have been approved qYes qNo
5. The Master Product Formula Record has all authorization signatures ? qYes qNo
6. The Master Manufacturing Batch Instructions has all authorization qYes qNo
signatures?
7. The manufacturing flow chart (identifying all equipment and process qYes qNo
parameters) is final with all authorization signatures?
10. Release Specifications (with narrower lower and upper limits) complete? qYes qNo
11. Check Specifications (with maximum lower and upper limits) complete? qYes qNo
14. The analytical methods and stability indicating assay are complete? qYes qNo
15. The PQ is not a regulatory requirement but a in-house dry run! qYes qNo
16. The Granulate Content Uniformity Protocol is prepared for qYes qNo
evaluation during PQ batch manufacture.
17. The Tablet Hardness Qualification protocol will be evaluated during the PQ qYes qNo
batch manufacture.
STANDARD OPERATING
PROCEDURES
CL # P-000-03-01Y2K Page 1 of 1
PROCESS QUALIFICATION
4
[End of Document]
Process
Qualification
‘these are the product specifications
indicating the product quality throughout the shelf life’
D
ifferent sampling Testing final blend content uniformity
techniques with the same
as a suitable in-process control may
thief can profoundly impact
well evaluate and highlight the
on sampling error, producing non
incoming ingredient batch-to-batch
re-presentative results, with
differences as well as the physical
respect to the true blend uniformity
variations in different lots of active
value.
material thus producing granule
♦ Sampling motion variation.
Bulk
♦ Sampling Angle
density and particle size of the
active material may be received within
♦ Sampling Orientation
the range specification, but at either
extremes of the specification limit.
♦ Bed depth However routine testing should not be a
GMP requirement as blending is not the
can bias results final unit process. Tablet compression
or capsule filling are the end
F actors that can bias the sample
processes!
in four different ways are:-
♦ Sampling motion (smooth, twisting Thus the necessity to establish both
jerking, or oscillating actions etc.) lower (LCL) and upper control limits
(UCL) for the content uniformity of the
♦ Sampling angle (vertical, acute, blend is self evident. The final blended
downwards). granulate assay should conform to
within the mean Ñ3 SD representing the
♦ Sampling thief orientation in the bed
(Side-sampling probe rotation of lower & upper control limits (See SOP).
chamber is up (3600á), down
0 0
(180 â), or on the side (either 45 ä
; 900 à or 1350 æ). References
♦ Depth of powder bed (top, middle or 1. Current Good manufacturing Practice of Certain
Requirements for the Finished Pharmaceuticals;
bottom sampling). Proposed Rules May 3 1996 (61 FR 20103).
In general, if development studies show 2. United States of America vs. Barr Laboratories
Inc., civil action for the district of N.J., Feb 1993.
the need for 3X sampling as a 3. J.T. Carstensen and M.V. Dali "Blending
prerequisite, then do so and amend Validation and Content Uniformity of low content
your SOPs (see model) as sampling …powder blends " Drug development and Industrial
Pharmacy Vol. 22 Issue 4 pp. 285-290 (1996).
error increases as the sample size 4.J Berman, A Schoeneman and JT Shelton, Unit
and/or formulation potency decreases. Dose Sampling - a tale of two thieves" Drug
development and Industrial Pharmacy Vol. 22 Issue
Final blending process, like Tablet 11 pp.1211-1132 (1996).
5. J Berman, and J.A. Planchard "Blend Uniformity
Hardness Qualification, is one of the and Unit Dose Sampling" Drug development and
series of unit processes that require Industrial Pharmacy Vol. 21 Issue 11 pp.1257-1283
(1995).
validation. This validation should be 6.J.T. Carstensen and C.T. Rhodes " Sampling in
performed at the Process Qualification Blending Validations" Drug development and
and / or Pivotal batch manufacturing Industrial Pharmacy Vol. 19 Issue 20 pp.2699-2708
(1993).
stage.
Final
Granulate
QUALIFYING
Tablet Hardness…
T ablet Hardness is an important
Qualification for a development
batch lot that has reached the
hardness range has been fully qualified
at the lower and upper hardness limits
on production machinery operating at
process qualification stage just prior to optimum target speeds - all performed
the pivotal manufacture. in a production environment.
During the commercial manufacture of Hardness Range Qualification means
a compressed tablet, the tabletting that, not only is tablet weight and
machines are set at an optimum thickness within specifications, but the
machine speed and compress at a set important dissolution profile remains
target hardness value. within the set parameters. More
This hardness value is developed from precisely the dissolution profile
experimental batch lots during the supports the specified Q value and
tablet development. Its value may be does not fluctuate beyond the lower or
close to the mean or average hardness the upper control limits (See Attached
range expressed between a lower and SOP of the hardness values (i.e. LCL
an upper hardness value. Such a range and UCL).
as NLT 8 - NMT16 Strong Cobb Units Normally only 10% of the batch is
(SCU) may be a common example, compressed at the lower and the upper
however ranges may be significantly hardness limits while the remaining
wider and still be acceptable such as 80% of the batch size is compressed at
NLT 6 - NMT 24 SCU, (Kg or SCU) the target hardness specifications.
may be used as the unit of Standard Pivotal batches produced for
measurement for hardness). regulatory submission are required to
deliver not less than 100 000 tablets
Hardness net (packed).
Its is expedient to produce a Pivotal
Qualification means batch on around 110 000 units - thus
that the dissolution the number of tablets run at the two
ends of the hardness limits would be
profile remains in approximately ten to eleven thousand
each.
specification Representative sampling during the
What makes these seemingly wide hardness qualification trials is
hardness range values reasonable and essential. Sufficient samples need to
appropriate for commercial be taken that fully represent the mini
manufacture is the fact that the tablet sub-lot for the standard routine quality
control,
TABLET
HARDNESS QUALIFYING TABLET HARDNESS
QUALIFICATION
retention samples and hardness
evaluation qualification tests. THE TABLET HARDNESS MAY BE
QUALIFIED DURING THE PIVOTAL
Prudent development labs will reserve RUN OR BEFORE DURING THE
a portion of the compressed tablets at Process Qualification Run
the hardness limit ends and place them
on real time stability.
It is well documented that tablet
Choose Upper & Lower Hardness
hardness may change with ageing, - values based on results from the
thus impacting on the dissolution Development PQ Batch
profile. This is especially the case
when the tablet moisture content may
be marginally higher (i.e. tablets
exceeding 2.0 to 2.5% LOD, or PREPARE THE WRITTEN PROTOCOL
greater). TITLED:
'TABLET HARDNESS QUALIFICATION'
Qualifying tablet hardness ranges
within the expected shelf life is at times
overlooked by the drug development COMPRESS BATCH
firm - an omission which may cause an IN THREE DISTINCT STAGES:-
out-of-specification product, while in 10% - LOW HARDNESS
80% - TARGET HARDNESS
the market place.
10% - HIGH HARDNESS
(Do not exceed a 15%-70%-15% ratio)
Qualify dissolution
profile on CAUTION
STANDARD OPERATING
SOP # D-000-02-089Y PROCEDURES
Page 1 of 2.
1. PURPOSE
The purpose of this Standard Operating Procedure is to provide a protocol (• ‚) to
qualify the recommended hardness range of [Immediate Release] [00] mg Tablets/
caplets, in order to achieve the dissolution profile which corresponds to the finished
product specifications
2. RESPONSIBILITY
Responsibility of actions is represented in the text by symbol:-.
• indicates Protocol is prepared by R&D Project and Process Development
Managers. Protocols are approved by the R&D and QA.
‚ indicates performed by validation team.
ƒ indicates performed by plant QA Technicians.
… indicates in-process testing performed by the plant QC Analytical Laboratory.
† indicates testing performed by the R&D Analytical Laboratory.
‡ indicates analysis and evaluation of the test data generated, performed by the
R&D Validation Team and approved by the R&D Quality Assurance.
3. FREQUENCY
Performed with the Process Qualification and the Pivotal Lot
4. PROCEDURE
PROSPECTIVE HARDNESS RANGE QUALIFICATION
4.1 Sampling Plan:
Samples are collected as they are compressed at target machine speed. The tablets/
caplets are compressed at the upper and lower limits of the hardness tablet
specification range. 10% of the batch is compressed at the upper and lower
hardness limits and 80% at the target limits.
Edition Number:
02
Effective Date
APPROVED
Ed. Status: DD/MM/YY ______________ __________ _______________ ______/__________
Supercedes: 01 Department R &D RA QC / QA
Handbook of Pharmaceutical Sect: 14.
14 11 Generic Development
ORAL TABLETS PROCESS QUALIFICATION CHAPTER 14
STANDARD OPERATING
SOP # D-000-02-089Y PROCEDURES
Page 2 of 2.
7.0 DOCUMENTATION
7.1 A Hardness Range Qualification Report (HRQR-‡) including tabulation of
results, statistical evaluation, process capability evaluation, report conclusions and
recommendations will be submitted to QA Unit, Production and Development Unit
Managers.
3
[End of Document]
Edition Number:
02
Effective Date
APPROVED
Ed. Status: DD/MM/YY ______________ __________ _______________ ______/__________
Supercedes: 01 Department R &D RA QC / QA
Handbook of Pharmaceutical Sect: 14.
14 12 Generic Development
TABLETS ORAL P I V O T A L CHAPTER 15
PIVOTAL
BATCH
‘the generic product has been developed and qualified
this is a key demonstration batch for regulatory submission.’
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
1. The product formula for the pivotal is the final marketing formula? qYes qNo
2. The Manufacturing Instructions are suitable for routine production? qYes qNo
3. The pivotal batch manufacturing equipment is standard production qYes qNo
equipment operated by production staff with routine QA personnel?
4. A side-by-side comparison of the pivotal equipment and the validation qYes qNo
batch equipment are similar, and differ in a change in scale only?
5. The pivotal batch production will follow all production SOPs? qYes qNo
6. The pivotal batch size is 10 % or greater of the largest proposed qYes qNo
commercial lot?
7. The complete pivotal batch must be 100 % filled and packaged in the qYes qNo
marketing container-closures (no part-packaging permitted)?
8. All production equipment has been physically checked for appropriate qYes qNo
recorders and control units as written in the pivotal documentation ?
9. The validation protocol for the first three full scale lots is drawn up? qYes qNo
10. The validation protocol addresses all key processing parameters, qYes qNo
that if changed, will significantly impact on product quality ?
11. All microbiological methodology has been audited and signed-off ? qYes qNo
12. Assays and test methods based on the USP, with in-house qYes qNo
modifications has been validated?
13. The active’s assay has been validated and is a stability indicating qYes qNo
test?
14. The stability protocol addresses the key stability indicating qYes qNo
specifications ?
15. The overall pivotal manufacturing file is audited and signed-off ? qYes qNo
STANDARD OPERATING
PROCEDURES
CL # P-HGD-03-01Y2K Page 1 of 1
4
[End of Document]
STANDARD OPERATING
SOP # P-000-05-01Y2K
PROCEDURES
Total Pages: 5
Page 1 of 5.
1. PURPOSE
The purpose of this Standard Operating Procedure is to describe the production in-
process sampling procedure and the testing to be performed on each pivotal lot or
pilot plant scale batch lot. This sampling plan is structured upon the FDA's
"Guidance on the Packaging of Test Batches", of February 8, 1995 Number 41-95" &
PDA Technical Report, April 30 1997. It meets the draft FDA June 1998 Guidance
For Industry, titled "Stability Testing of Drug Substances and Drug Products".
2. RESPONSIBILITY
Responsibility of actions is represented by the relevant symbol in the text.
• Symbol indicates Protocol is prepared by Development Project Unit and Process
Development Managers. Protocols are approved by the Development and QA Unit.
‚ Symbol indicates performed by process validation team.
ƒ Symbol indicates sampling performed by the Plant QA Technicians.
„ Symbol indicates In-process testing performed by the Plant QA Technicians.
… Symbol indicates Batch Release Testing performed by the Plant QC Analytical
Laboratory.
† Symbol indicates testing performed by the Development Analytical Laboratory.
‡ Symbol indicates analysis and evaluation of the test data generated, performed by
the Development Unit Validation Team and approved by the Development Quality
Assurance Unit.
3. FREQUENCY
The procedure is performed with each pivotal or pilot-plant batch lot.
4. PROCEDURE
4.1 In-Process Control - Sampling and Testing
Final Blended Material - (Immediately prior to compression or capsule filling).
Physical Testing - Sampling Protocol
A total of Six samples (about 100g per sample) is collected from the storage
containers, representing the top, middle and the end of the final blended material.
Three 100g samples are used for testing and the balance of the samples reserved
for further testing, if so required.
STANDARD OPERATING
SOP # P-000-05-01Y2K
PROCEDURES
Total Pages: 5
Page 2 of 5.
Chemical Testing - Sampling Protocol - (continued)
Where sampling procedures from the Y-Cone Twin shell blender are not possible,
the reasons are described in the written product protocol.
In the case of a flow-bin being used is as a blender, then the samples are collected
from the top, middle and bottom of the blending unit.
Sample collection ‚ƒ ƒ
40 samples are collected
10 samples, each equivalent to the approximate weight of one (to three)³ unit dose(s)
‚ƒ, are collected from the storage containers, according to SOP, "Sampling of
Granulate for Blend Uniformity". (³ See limits and Limitations)
30 samples will be collected ‚ƒ ƒ as above and stored as retention samples for
additional testing, if required. When a flow-bin is used both as a blending unit and
as a storage container, no additional samples are taken.
Sampling collection:
The samples are collected via an appropriate “Sampling Thief” equipped with the
necessary die - unless otherwise instructed in the written protocol.
4.2 Physical Testing Protocol
Each sample is tested for:
• Sieve analysis (wet granulation process only)
• Bulk and tapped density
4.3 Chemical Testing Protocol
• The samples will be assayed according to the full monograph.
4.4 Finished Processed Material - Tablets and Capsules
Applies to compressed Tablets, Caplets and Filled Capsules and Hardness Range
Qualification Testing for tablets and tablet cores only.
Sampling Protocol
Samples are collected at the target compression speed at the upper and lower limits
of hardness specification.
Testing Protocol
Tablets compressed at the upper and lower limit will be tested as follows [¹ Tests are
performed on the same tablets in the following order.]
• 20 tablets for thickness ¹
• 20 tablets for weight ¹
• 20 tablets for hardness ¹
STANDARD OPERATING
SOP # P-000-05-01Y2K
PROCEDURES
Total Pages: 5
Page 3 of 5.
Testing Protocol †
Each sample will be tested † as follows - ¹ Tests performed on the same tablets:
• 20 tablets for thickness ¹
• 20 tablets/capsules for weight ¹
• 20 tablets for hardness ¹
For Friability:-
• For tablets less than 650 mg, test 6.0-6.5 g of tablets, but not less than 20
tablets.
• For tablets more than 650 mg, test 10 tablets.
For Tablet / Capsule Dissolution Profile:-
• Tablets / capsules ²
• Assay on each sample as per product monograph.
² Where tablets are film-coated, a dissolution profile is performed on tablets collected at each of the
three exit time interval, making a total of 18 tablets.
STANDARD OPERATING
SOP # P-000-05-01Y2K
PROCEDURES
Total Pages: 5
Page 4 of 5.
STANDARD OPERATING
SOP # P-000-05-01Y2K
PROCEDURES
Total Pages: 5
Page 5 of 5.
This SOP is restricted to the sampling and testing of in-process controls during
pivotal or pilot-scale batch manufacture for chemical and physical testing, and
includes Tablet Hardness Qualification Testing. This SOP does not specify the
actual number of packed units to be representatively sampled for: QC Retention,
Stability Profile, Stability Reserve or Bioequivalence Testing and Bioequivalence
Reserve.
³ Sample size may be increased to three dosage units where appropriately qualified
in the process development report.
4 In-process
Granule Content Testing (Content Uniformity):
Where appropriately qualified by suitable batch analysis (or clearly established
during process development), the in-process lower and upper limits for Granule
Content Testing (Content Uniformity) may be narrowed to:
The Upper control limit (UCL) is defined as the mean + 2 x Standard Deviation
The Lower control limit (LCL) is defined as the mean - 2 x Standard Deviation
7. CORRECTIVE ACTION
Out-of-specification test results (OOS) are handled according to the firms current
Out-of-Specification SOP.
8. DOCUMENTATION
Each protocol will be accompanied • by a manufacturing process flowchart.
The sampling record ‚ forms are filed with the manufacturing batch records.
Sampling Record Forms for data completion at the time of sampling are as follows:
Attachment 1 - " Sampling Record for Final Blended Granulate "
Attachment 2 - " Sampling Record for compressed Tablet / Caplet & Capsule Filling"
Attachment 3 - " Sampling Record for Coated Tablets / Caplets "
Attachment 4 - " Sampling Record for Finished Product Release & Stability T0 Test "
3
[End of Document]
Auditing the
Inspecting and comparing the pivotal
batch documentation line-for-line and
page-by-page is a prudent, worthwhile
Pivotal Batch and judicious auditing operation.
Retrieving and reviewing the plant
production and cleaning logs or
T he Pivotal (Regulatory or Exhibition)
Batch is the demonstration batch lot
that is submitted to the regulators
ingredient inventory cards are a
necessary part of the job. Checking
as the example and legal basis on batch numbers, dates and signatures,
which the future commercial lots will be possible transposed numbers or
marketed and sold. It represents a omitted data, require careful vigilance
baseline of procedures and product by the auditor or audit team. Auditing
specifications that are the regulatory with a formal procedure, simplifies the
basis for government agency approval. task and allows the audit checklist to
grow when new and unusual errors or
This is generally the first production omissions are detected.
batch in which all the production and
control parameters and specifications Update audit checklists
have been fully set. Little practical
experience has been gained so far, after each pivotal lot
from the manufacture of a minimum 100 An audit checklist that remain static
000 units (net / packed quantity). over the years, may well reflect an
Production familiarity, experience and ineffective inspection, analysis and
process fine tuning, by the firm will be detection procedure.
gauged, only after the product has been The Pivotal Batch Audit Checklist.
approved, during the many commercial Checklist Do's:-
production manufacturing lots.
♦ Divide audit checklist into
Error-Free Pivotal representative sections portraying each
documentation saves operation, manufacturing and control
procedure.
time and money ♦ Add new check points to the list,
when a new error or omission arises.
Thus the need to get it right, at the very ♦ Test the checklist, by evaluating
first attempt at the production level routine production batches, to get a
(pivotal lot) is paramount. Careless sense of possible omission categories.
omissions in the batch documentation ♦ Break multiple check points into
will eventually result in an agency individual audit items on the checklist.
deficiency letter and thereby delaying
approval for an additional few months -
♦ Carefully cross-reference hand-
written dates and times on the
costing both time and valuable
production forms against machine print-
development and lost sales dollars.
outs that have immediately follow the
Experienced Pharmaceutical Firms operational step.
know the high costs of undetected ♦ Remember - weighing, LOD, HPLC
documentation omissions or data fields, and temperature print-outs and charts
and have set up efficient development normally give the date and the exact
quality assurance units that closely time of the operation or test procedure.
examine, inspect, audit and review Evaluate production yields and
every aspect of the batch adherence to time-limitation against
documentation for submission actual weighing & recording print-outs. 2
STANDARD OPERATING
SOP # P-000-02-01Y2K
PROCEDURES
STANDARD OPERATING
SOP # P-000-02-01Y2K
PROCEDURES
STANDARD OPERATING
SOP # P-000-02-01Y2K
PROCEDURES
Generic Bioequivalence
and the Reference Listed Drug
Bioavailability Bioequivalence
COMPARATIVE DISSOLUTION PROFILE FOR DRUG PRODUCT
LOTS USED IN BIOEQUIVALENCE STUDIES:
[Generic Name] Tablets USP [000.0] mg. Lot No:1234
12 TABLETS
100
80
60 P-01234
RLD AA0000
% DISSOLVED 40
20
10 20 30 40 50
TIME (minutes)
Bioavailability / Bioequivalence
COMPARATIVE DISSOLUTION PROFILE FOR DRUG PRODUCT
LOTS USED IN BIOEQUIVALENCE STUDIES:
12 Tablets per test on multiple RLD batch lots under normal and stressed conditions
Stress Study
3 months Stability at 40° C / 75% RH
Figure No. 2. Comparative Dissolution Profile
120
100
80
P-01234
60 RLD-AA000
% DISSOLVED
40
Time Scale changes
20 for different active
ingredients i.e.
0 generally from
10 20 30 40 50 60 30 - 60 minutes
TIME (minutes)
♦ [Generic name] Tablets [USP] [000.0] mg. Lot: 1234 CoA No:
0000
♦ [RLD] Tablets [USP] [000.0] mg. Lot: AA000 CoA No: 0000
The analytical results of the Certificates of Analysis for [Generic Company Name Inc. / Ltd.] and
[RLD Company Name Inc. / Ltd.] Drug Product lots were tested the Analytical Research Laboratories
of [Generic Company Name Inc. / Ltd. & Address].
MODIFIED RELEASE
DESIGN OF STUDY
CROSS OVER [Most Drugs]
PARALLEL [Long ½ life Drugs]
BOTH
BIOSTUDIES
0.80 1.0 1.25 PASS IBE
but
TEST / REFERENCE RATIO FAIL ABE
Bioequivalence Failure
Bioequivalence Pass
With acknowledgments:
Fast marketing Authorization for Generics London Kensington Posthouse Conference December 3&4th 1997;
Understanding Bioequivalence and Therapeutic Equivalence Conference June 25/26 1998 Rockville MD USA.
Professor Laszlo Endrinyi - University of Toronto; Walter W Hauck PhD., Thomas Jefferson University. Andrew
Grieve-Biometrics Department Pfizer Central Research; Anderson and Hauck J. Pharmacokin. & Biopharm.
1990. Shein-Chung Chow & Jen-Pei Liu (1992) Design and Analysis of Bioavailability and Bioequivalent
Studies, Marcel Dekker; Drug Information Journal 03, July-Sept 1995, Dedicated to Bioavailability and
Bioequivalence.
Individual bioequivalence's (IBE) basic tenet assumes that the currently employed
methods and criteria for the assessment of bioequivalence are inadequate to
provide assurance that the brand product can be switched to a generic product and
vice versa - however…
…the demands for an equivalent of the generic should not exceed those required to
show equivalence of the brand itself. New requirements should be based on a
demonstrated need for change or a bona fide risk and not on conjecture or
theoretical supposition.
Switchability Term DIFFERENCE
DIFFERENCE OF VARIANCES
OF MEANS
Subject by
formulation
interaction
(s-b-f)
µT - µR)2 +
(µ σD2 + σ2WT _ σ2WR) ≤ θP
(σ
σ2W0
POSITIVE ASPECTS REGULATORY
Addresses switchability CONSTANT SCALED CRITERIA
but at a study cost of (UN-SCALED) Ln 1.25
more than double
average bioequivalence
POSITIVE ASPECTS
Does not assume that the
variability of the Test and MAY BE SCALED
Reference formulations are the FOR HVDS
same - therefore rewards a better
manufactured product σ2WR
Individual bioequivalence (IBE) basic tenet assumes that if one were to ask a man
or woman in the street what they believe is meant by approval of a generic drug as
equivalent to a name-brand product…
… the answer will come back something like this, " it doesn't really matter which
product I take"
Switchability Term WITHIN SUBJECT
DIFFERENCE (Not always significant) VARIANCES
OF MEANS
(AUC; Tmax)
Subject by
formulation
interaction
(s-b-f)
µT - µR)2 +
(µ σD2 + σ2WT _ σ2WR) ≤ θP
(σ
σ2WR
REGULATORY
NEGATIVE ASPECTS
CRITERIA SET
Switchability (s-b-f SCALED
variation) not significant Ln 1.25
Highly Variable Drugs
in many studies with
healthy subjects
NEGATIVE ASPECTS Scaling helps highly
More variability of σ2WR will variable drugs (HVD) to
make it easier to pass pass the individual
bioequivalence criteria
OUTLINE IN VIVO BIOSTUDY for should be able to capture the Cmax and Tmax
during the absorption period. Sampling
AVERAGE BIOEQUIVALENCE should be carried out for at least three
FASTED STUDY - OVERALL PICTURE
terminal elimination half-lives for both parent
The Purpose of this general model outline of drug and active metabolite(s). Whole blood,
an in vivo bioequivalence study is intended plasma or serum, whichever is appropriate
as a BASIC guide. for the analytes, should be harvested
The design of the actual study may vary promptly and samples should be frozen at -
depending on the drug and dosage form. 20oC or -70oC to maintain sample stability.
Objective - To compare the rate and extent
of absorption of the Test Drug Product for Analytical Method - The assay
which bioequivalence is sought to the methodology selected should ensure
Reference Listed Drug (RLD). specificity, accuracy, interday and intraday
Design - The study design should be a precision, linearity of standard curves, and
single dose, two-treatment, two-period adequate sensitivity, recovery, and stability
crossover with adequate washout period of the samples under the storage and
between the two phases of the study. Equal handling conditions associated with the
numbers of subjects should be randomly analytical method.
assigned to each of the two dosing Pharmacokinetic Analysis - From the
sequences. plasma drug concentration-time data, AUC0-t,
Selection of Subjects: - The number of AUC0-inf, Cmax, Tmax, Kel and t1/2 should be
subjects enrolled in the bioequivalence study estimated.
should be determined statistically to account
for the intra-subject variability and to meet Statistical Analysis - Analysis of variance
the current bioequivalence interval. appropriate for a crossover design on the
pharmacokinetic parameters using the
Procedure: - Each subject should receive general linear models procedures of SAS or
the following two treatments: an equivalent program should be performed,
Treatment 1: Generic Product or Test Drug. with examination of period, sequence and
Treatment 2: Reference Listed Drug (RLD). treatment effects. The 90% confidence
Following an overnight fast of at least 10 intervals for the estimates of the difference
hours, subjects should receive either between the test and reference least
Treatments 1 or 2 above with 240 mL water. squares means for the pharmacokinetic
Food should not be allowed until 4 hours parameters (AUC0-t, AUC0-inf, Cmax) should
after dosing. Water may be allowed after be calculated, using the two one-sided t-test
the first hour. Subjects should be served procedure.
standardized meals beginning at 4 hours
during the study.
REFERENCES
Restrictions - Prior to and during each study 1. Code of Federal Regulations 210.3(b)(2) and (10), 310.3(b)
phase, water may be allowed ad libitum and (g), and 320.1(a) and (e).
except for 1 hour before and after drug 2. Federal Register. Vol. 59, No. 183, September 22, 1994, pp
48754-59.
administration. The subject should be 3. “Guideline for Industry: Stability Testing of New Drug
served standardized meals and beverages at Substances and Products,” U.S. Department of Health and
specified times. No alcohol or xanthine- or Human Services, FDA, September 1994.
caffeine-containing foods and beverages 4. Guideline for Submitting Documentation for the Manufacture
and Controls for Drug Products,” U.S. Depart-ment of Health and
should be consumed for 48 hours prior to Human Services, FDA, February 1987.
each study period and until after the last 5. Policy and Procedure Guide #22-90: “Interim Policy on
blood sample is collected. Exceptions to the Batch-Size and Production Condition
Requirements for Non-Antibiotic, Solid, Oral-Dosage Form Drug
Blood Sampling: - Blood samples should be Products Supporting Proposed ANDA’s”, U.S. Department of
Health and Human Services, Center for Drug Evaluation and
collected in sufficient volume for analysis of Research, Office of Generic Drugs, September 13, 1990.
parent drug and active metabolite(s), if any.
The sampling times should be such that it
Where possible age or stress one of Reference Listed Drug batch Lots
the RLD lots and evaluate with and placed on accelerated stability at 40° C
without a surfactant. / 75% RH for 3 - 6 months. (Evaluate
Assay and Impurity profiles).
Evaluate profiles at low and high rpm
rotational speeds. The higher cost of a
It is significant to evaluate the
full comparative dissolution program variability in the RLD product under
may well be the difference between a normal and, after stressed conditions
'biostudy' pass or failure. as well as at different RLD
manufacturing dates (where the RLD is
Dissolution Profile 'Similarity Test'
purchased with different Lot Numbers
calculate the unbiased 'Similarity
i.e. made at different times).
Factor' for each CDP. The result ƒ² Success of a costly Bioequivalence
should be between 50 to 100 to study may hinge or depend on the
demonstrate 'similarity' between the detail of the RLD's data (i.e. Multiple
two formulations. Where similarity is lots at different manufacturing date,
obtained cross check values using normal, and aged studies).
alternative functions (e,g Chow, Intra-batch and inter-batch variability of
ANCOVA etc.) the RLD should be evaluated with care.
♦ Bioequivalence Study - Pilot
In the case of high cost biostudies a
pilot study may give meaningful results
DISSOLUTION STUDIES
What is Important?
and enable necessary formula or
process adjustments in intricate drug
comparisons. Should the pilot study fail Fully Functioning Equipment
Routine Calibration & Performance Verification.
or 'just' fail' - it is important to note the No minor mechanical damage or misalignments.
full participant study would have failed
and at a significantly higher cost. In
'just fail' situations, reformulate drug Fully Validated
product and in parallel improve the Dissolution Assay Procedures available
before Formula & Process Optimization,
discrimination power of the dissolution PQ, and Pivotal Lots.
testing procedure.
♦ Bioequivalence Study - Full Discriminating Test Procedures
Agitation, pH, Surfactant, Media, Deaeration
Your R&D unit should be ready to
perform a full-scale biostudy supported
with adequate CDP data and statistical Operator Excellence & Training
assessments to pass the pending Routine Standard Operating
biostudy against the Reference Drug. Procedures
prepare
SLOW
MEDIUM
FAST profiles Optimum Formula
Dissolution Testing of IR
Solid Oral Dosage Forms
FDA's DISSOLUTION TESTING PROCEDURES
SOLID ORAL DOSAGE FORMS
I. INTRODUCTION II. BACKGROUND
The BCS suggests that for high consistency and to signal potential
solubility, high permeability (case 1) problems with in vivo bioavailability. For
drugs and in some instances for high NDAs, the dissolution specifications
solubility, low permeability (case 3) should be based on acceptable clinical,
drugs, 85% dissolution in 0.1N HCl in pivotal bioavailability, and/or
15 minutes can ensure that the bioequivalence batches.
bioavailability of the drug is not limited For ANDAs/AADAs, the dissolution
by dissolution. In these cases, the rate specifications should be based on the
limiting step for drug absorption is performance of acceptable
gastric emptying. bioequivalence batches of the drug
The mean T50% gastric residence product.
(emptying) time is 15-20 minutes under The NDA dissolution specifications
fasting conditions. Based on this should be based on experience gained
information, a conservative conclusion during the drug development process
is that a drug product undergoing 85% and the in vitro performance of
dissolution in 15 minutes under mild appropriate test batches. In the case of
dissolution test conditions in 0.1N HCl a generic drug product, the dissolution
behaves like a solution and generally specifications are generally the same as
should not have any bioavailability the reference listed drug (RLD).
problems. The specifications are confirmed by
If the dissolution is slower than gastric testing the dissolution performance of
emptying, a dissolution profile with the generic drug product from an
multiple time points in multimedia is acceptable bioequivalence study.
recommended.
If the dissolution of the generic product
In the case of low solubility / high is substantially different compared to
permeability drugs (case 2), drug that of the reference listed drug and the
dissolution may be the rate limiting step in vivo data remain acceptable, a
for drug absorption and an IVIVC may different dissolution specification for the
be expected. generic product may be set.
A dissolution profile in multiple media is Once a dissolution specification is set,
recommended for drug products in this the drug product should comply with
category. In the case of high solubility / that specification throughout its shelf
low permeability drugs (case 3), life.
permeability is the rate controlling step
and a limited IVIVC may be possible,
The International Conference on
harmonization (ICH) Q1A guideline
depending on the relative rates of
(Stability Testing of New Drug
dissolution and intestinal transit.
Substances and Drug Products) has
Drugs in case 4 LS/LP (i.e., low recommended that for an NDA, three
solubility / low permeability drugs) batches (two pilot and one smaller
present significant problems for oral scale) be placed into stability testing.
drug delivery.
These batches also may be used to set
Ø SETTING DISSOLUTION dissolution specifications when a
suitable bioequivalence relationship
SPECIFICATIONS × exists between these batches and both
Invitro dissolution specifications are the pivotal clinical trial batch and the
established to ensure batch-to-batch drug product intended for the market.
Three key categories of dissolution test pharmaceutics and CMC review staff in
specifications for immediate release the Office of Pharmaceutical Science
drug products are the following. (OPS).
Ü SINGLE-POINT SPECIFICATIONS For NDAs, the specifications should be
As a routine quality control test. (For based on the dissolution characteristics
highly soluble and rapidly dissolving of batches used in pivotal clinical trials
drug products.) and/or in confirmatory bioavailability
studies.
Ü TWO-POINT SPECIFICATIONS If the formulation intended for marketing
1. For characterizing the quality of the
differs significantly from the drug
drug product.
product used in pivotal clinical trials,
2. As a routine quality control test for
dissolution and bioequivalence testing
certain types of drug products (e.g.,
between the two formulations are
slow dissolving or poorly water soluble
recommended.
drug product like carbamazepine).
Ü DISSOLUTION PROFILE COMPARISON Dissolution testing should be carried
1. For accepting product sameness out under mild test conditions, basket
under SUPAC-related changes. method at 50/100 rpm or paddle method
2. To waive bioequivalence at 50/75 rpm, at 15-minute intervals, to
requirements for lower strengths of a generate a dissolution profile. For
dosage form. rapidly dissolving products, generation
3. To support waivers for other of an adequate profile sampling at 5- or
bioequivalence requirements. 10-minute intervals may be necessary.
In the future, a two-time point approach For highly soluble and rapidly
may be useful, both to characterize a dissolving drug products (BCS classes
drug product and to serve as quality 1 and 3), a single-point dissolution
control specification. test specification of NLT 85% (Q=80%)
in 60 minutes or less is sufficient as a
Approaches for Setting routine quality control test for batch-to-
Dissolution Specifications for batch uniformity.
a New Chemical Entity For slowly dissolving or poorly water
Dissolution methodology and soluble drugs (BCS class 2), a two-
specifications developed by a sponsor point dissolution specification, one at
are presented in the bio-pharmaceutics 15 minutes to include a dissolution
section (21 CFR 320.24(b)(5)), and the range (a dissolution window) and the
chemistry, manufacturing, and controls other at a later point (30, 45, or 60
section (21 CFR 314.50(d)(1)(ii)(a)) of an minutes) to ensure 85% dissolution, is
NDA. recommended to characterize the
quality of the product.
The dissolution characteristics of the
drug product should be developed The product is expected to comply with
based on consideration of the pH dissolution specifications throughout its
solubility profile and pKa of the drug shelf life. If the dissolution
substance. characteristics of the drug product
change with time, whether or not the
The drug permeability or octanol / water specifications should be altered will
partition coefficient measurement may depend on demonstrating
be useful in selecting the dissolution bioequivalence of the changed product
methodology and specifications. The to the original biobatch or pivotal batch.
dissolution specifications are
established in consultation with bio-
assess batch-to-batch product quality If the products with the extreme range
provided the bioequivalence is of dissolution characteristics are found
maintained. to be bioequivalent to the standard or
Recent examples involving soft and hard gelatin
capsules show a decrease in the dissolution
the to be marketed dosage form, future
profile over time either in SGF or in SIF without batches with dissolution characteristics
enzymes. This has been attributed to pellicle between these ranges should be
formation. When the dissolution of aged or equivalent to one another.
slower releasing capsules was carried out in the
presence of an enzyme (pepsin in SGF or This approach can be viewed as
pancreatin in SIF), a significant increase in the verifying the limits of the dissolution
dissolution was observed. In this setting, specifications. Product dissolution
multiple dissolution media may be necessary to
adequately assess product quality.
specifications established using a
mapping approach will provide
Mapping or Response Surface maximum likelihood of ensuring stable
Methodology quality and product performance.
Mapping is defined as a process for Depending on the number of products
determining the relationship between evaluated, the mapping study can
Critical Manufacturing Variables (CMV) provide information on invitro-invivo
and a response surface derived from an correlations and/or a rank order
in vitro dissolution profile and an in vivo relationship between invivo and invitro
bioavailability data set. The CMV data.
include changes in the formulation,
process, equipment, materials, and
Invivo-Invitro Correlations
methods for the drug product that can For highly water soluble (BCS Class 1
significantly affect in vitro dissolution and 3) immediate release products
The goal is to develop product using currently available excipients and
specifications that will ensure manufacturing technology, an IVIVC
bioequivalence of future batches may not be possible.
prepared within the limits of acceptable For poorly water soluble products, BCS
dissolution specifications. Several class 2, an IVIVC may be possible.
experimental designs are available to The value of dissolution as a quality
study the influence of CMV on product control tool for predicting invivo
performance. performance of a drug product is
One approach to study and evaluate significantly enhanced if an invitro-in
the mapping process includes:- vivo relationship (correlation) is
established. The in vitro test serves as
(1) prepare two or more dosage a tool to distinguish between acceptable
formulations using CMV to study their in and unacceptable drug products.
vitro dissolution characteristics
Acceptable products are bioequivalent,
(2) test the products with fastest and in terms of invivo performance, whereas
slowest dissolution characteristics along unacceptable products are not. To
with the standard or the to be marketed achieve an invitro-invivo correlation, at
dosage form in small groups (e.g., least three batches that differ in the in
n>12) of human subjects vivo as well as the in vitro performance
(3) determine the bioavailability of the should be available.
products and in vitro-in vivo If the batches show differences in invivo
relationship.
performance, then invitro test conditions
The products with extreme dissolution can be modified to correspond with the
characteristics are also referred to as in vivo data to achieve an invitro-invivo
side batches (Siewert 1995). correlation.
USE Deaerate
Calibrate a VALIDATED the
the System Dissolution dissolution
FULLY Assay medium
Dissolution Validation
Use mild rpm
Mimic the pH
conditions
Paddle 50-75 rpm of the GIT
Basket 50-100 rpm Target Site
Dissolution Performance
Sample every Use a Surfactant
15 minutes for for water insoluble or
IR Dosages sparing soluble drugs
(i.e. Na lauryl sulfate)
Dissolution Comparisons
Perform a
Comparative Evaluate the Choose the
Dissolution SIMILARITY right statistical
Profile Statistical Approach
(4 or more points) Factors
Dependent Independent
Approach Approach
DRUG DEVELOPMENT
Because of the critical nature of the first Acceptable bioequivalence data and
two of these steps, invitro dissolution comparable invitro dissolution and CMC
may be relevant to the prediction of data are required for approval of
invivo performance. abbreviated new drug applications
(ANDAs) (21 CFR 314.94).
When 85% of The invitro specifications for generic
Highly Soluble Drugs products should be established based
dissolve in 15 minutes on a dissolution profile.
The BCS applies primarily to immediate bioavailability of the drug is not limited
by dissolution.
release drug preparations but impacts
on HS / LP for Controlled Release Oral Bioequivalent studies are
preparations. generally performed on
PERMEABILITY & SOLUBILITY TABLE Highest Dosage Strength
Case 1: High Solubility - High In these cases, the rate limiting step for
Permeability Drugs drug absorption is gastric emptying.
Case 2: Low Solubility - High The mean T50% gastric residence
Permeability Drugs (emptying) time is 15-20 minutes under
Case 3: High Solubility - Low fasting conditions.
Based on this information, a
Permeability Drugs
conservative conclusion is that a drug
Case 4: Low Solubility - Low product undergoing 85% dissolution in
Permeability Drugs 15 minutes under mild dissolution test
conditions in 0.1N HCl behaves like a
This classification can be used as a solution and generally should not have
basis for setting in vitro dissolution any bioavailability problems.
specifications and can also provide a
basis for predicting the likelihood of Soluble Drugs dissolving
achieving a successful in vivo-in vitro over 85% in less than 15 min
correlation (IVIVC). in acid media
Highly Soluble Drugs can be considered as a liquid
are defined as:- preparation
Highest Strength dissolving If the dissolution is slower than gastric
in <250 mL buffer emptying, a dissolution profile with
multiple time points in multimedia is
The solubility of a drug is determined by recommended.
dissolving the highest unit dose of the In the case of low solubility / high
drug in 250 mL of buffer adjusted permeability drugs (case 2), drug
between pH 1.0 and 8.0. dissolution may be the rate limiting step
A drug substance is considered highly for drug absorption and an IVIVC may
soluble when the dose/solubility volume be expected.
of solution are ≤250 mL. A dissolution profile in multiple media is
High-permeability drugs are generally recommended for drug products in this
those with an extent of absorption that is category. In the case of high solubility /
greater than 90% in the absence of low permeability drugs (case 3),
documented instability in the permeability is the rate controlling step
gastrointestinal tract or those whose and a limited IVIVC may be possible,
permeability has been determined depending on the relative rates of
experimentally. dissolution and intestinal transit.
The BCS suggests that for high Drugs in case 4 LS/LP (i.e., low
solubility, high permeability (case 1) solubility / low permeability drugs)
drugs and in some instances for high present significant problems for oral
solubility, low permeability (case 3) drug delivery.
drugs, 85% dissolution in 0.1N HCl in 15
minutes can ensure that the
BIOPHARMACEUTICS
CLASSIFICATION SYSTEM (BCS)
The Biopharmaceutics Classification System (BCS) designed primary for IR drugs,
projects possible Invitro-Invivo Correlations between the drug classification (Solubility /
Permeability) and dissolution parameters of the Generic and Reference Drug. Where
appropriate, in-vitro dissolution tests involving single/multipoint dissolution profiles in
varying media may be used to demonstrate similarity between reference and generic
product. The BCS applies primarily to immediate release drug preparations but impacts
on HS / LP for CR preparations
Table 1:
GROUP Drug Dissolution IVIVC
Solubility EXPECTATION
Parameters Studies needed
Permeability
Classification System
Draft Guidance to Industry
"It is neither feasible nor desirable that active moieties / active ingredients that
studies of bioavailability be conducted are highly soluble and highly permeable
for all drugs or drug products. Certain from other drug products.
classes of drugs for which evidence of For such products, in vivo
bioequivalence is critical should be demonstration of bioequivalence may
identified. Selection of these classes of not be necessary because the BA/BE of
drugs should be based on clinical a drug product so characterized
importance, ratios of therapeutic to toxic approaches that of a solution and is thus
concentrations in blood, and certain self-evident (21 CFR 320.22(b) (3)).
pharmaceutical characteristics"
Furthermore, a suitable invitro / Invivo
Based on this and other
correlation can be assumed for a rapidly
recommendations of the panel, FDA dissolving drug product of a highly
proposed and finalized regulations in soluble and highly permeable drug
1977 entitled: substance, as long as its inactive
Bioequivalence Requirements and In ingredients do not significantly affect
Vivo Bioavailability Procedures absorption of the active ingredients.
(42 FR 1624; January 7, 1977). Conversely, drugs that are poorly
These regulations, which evolved over permeable, poorly soluble, and/or
time and are now codified at [21 CFR Part formulated in slowly dissolving dosage
320.33,] provide criteria for assessing forms may be considered to be drugs
actual or potential bioequivalence with actual or potential BE problems.
problems.
III. THE BIOPHARMACEUTICS
DRAFT
For drug products that were considered
to pose bioequivalence problems, the
regulations required that BA/BE be
demonstrated through in vivo studies.
CLASSIFICATION SYSTEM
The Biopharmaceutics Classification
System (BCS) is a general approach
GUIDELINE
For drug products that were not
considered to pose bioequivalence
problems, BA/BE could be demonstrated
that can be used by sponsors and
applicants to justify a biowaiver for
immediate release (IR) solid oral dosage
forms.
through in vitro studies.
The approach is based on the fact that
At the time of the passage of the Drug
invivo dissolution differences in the
Price Competition and Patent Term gastrointestinal tract are a primary
Restoration Act of 1984 (Waxman- reason for observed differences in
Hatch), FDA’s policy was to require bioavailabilities of two IR products
invivo demonstration of bio-equivalence
containing the same drug substance.
for all post-1962 (post-DESI2) non-
solution drug products. With occasional In the BCS, a drug is classified as
exceptions, this approach has continued belonging to either:-
to the present. ¯ 1) high or low solubility class,
PURPOSE ¯ 2) a high or low permeability class,
The purpose of this guidance is to ¯ 3) a IR dosage form is categorized
describe alternative ways, not based on as belonging to a rapid or slow
invivo methods, acceptable to FDA for dissolving class.
demonstrating bioequivalence for certain These classes and methods for
post-1962 immediate release solid oral classifying a drug are discussed in the
drug products based on a classification following sections. The term class
system that distinguishes rapidly boundary is used to indicate how the
dissolving drug products containing class should defined.
administration
protocols.
DRAFT
minimum volume anticipated in the
stomach at the time of drug
during the study
IV. METHODOLOGY
CLASSIFYING A DRUG
FOR
to DRAFT
A low permeability internal standard is
suggested ensure intestinal
membrane integrity. The permeability
based on the BCS should contain
documentation on the following:
¯ 1. The drug substance for which a
GUIDELINE
values of the two internal standards can
be used to verify reproducibility of the
experimental method.
waiver is being requested should be
highly soluble and highly permeable, as
defined above.
¯ 2. An IR drug product should be
The internal standards should be
compatible with the drug being rapidly dissolving, as defined above.
evaluated (i.e., they should exhibit no ¯ 3. For waiver of an in vivo BA study,
physical or chemical interactions). dissolution should be greater than 85%
A list of potential model drugs and in 30 minutes in the three recommended
chemicals along with their permeability dissolution media.
class membership is provided in Tablet For waiver of bioequivalence, test and
A. reference products should exhibit similar
In vitro methods, such as those using a dissolution profiles under the dissolution
cultured monolayer of human intestinal test conditions defined for rapidly
cells, should be further examined to dissolving products.
identify any expressions of specialized Two dissolution profiles may be
transport/efflux systems for the selected considered similar when compared using
experimental conditions (e.g., the f2 metric (f2 ≥50) as described in the
bidirectional transport studies using guidance for industry on dissolution
model compounds, such as verapamil, testing4.
of the p-glycoprotein efflux system). When both the test and the reference
Permeability class membership products dissolve 85% or more of the
determined using such in vitro methods label amount in ≤15 minutes, in all three
can be considered reliable when:- dissolution media recommended above,
a profile comparison is unnecessary.
DRAFT
VI. ADDITIONAL CONSIDERATIONS
WHEN PLANNING A REQUEST FOR A
WAIVER
highly permeable drug that is formulated
in a rapidly dissolving IR product.
When new excipients, or atypically
When
GUIDELINE
requesting a waiver for in vivo
BA/BE studies for IR solid oral dosage
forms, applicants also should consider
the following issues, which could affect
large amounts of commonly used
excipients, are used in an IR dosage
form, additional information documenting
the absence of an impact on
their request or the documentation of bioavailability could be requested by the
their request. Agency.
A. Instability in the Gastrointestinal Include a list of
Tract
Determining the extent of absorption in
formula excipients
humans based on mass balance using when requesting the
total radioactivity in urine does not Biowaver
consider the extent of degradation of a
drug in the gastrointestinal fluids prior to Such information can be supplied via a
intestinal membrane permeation. relative bioavailability study using a
Also, some methods for determining simple aqueous solution as the
reference product.
permeability could be based on loss, or
clearance, of a drug from fluids perfused A request for biowaiver based on the
into the human and/or animal BCS should include a list of all
gastrointestinal tract either in vivo or ex excipients used in the products, the
vivo. Documenting that drug loss from amount used in the test product,
the gastrointestinal tract arose from intended functions, a brief summary
intestinal membrane permeation, rather describing the manufacturing process,
than a degradation process, will help and a list of equipment used.
ATTACHMENT A: References:
SUGGESTED MODEL DRUGS 1 DESI2 drugs were identified as part of the Drug
Efficacy Study Implementation Program, which was a
Suggested model drugs for use in retrospective evaluation of the effectiveness of drugs
cleared through the new drug procedures only on the
establishing method suitability as basis of safety between 1938 and 1962.
described in section IV.
The DESI program was undertaken to implement the
The permeability class memberships of requirement set forth in the 1962 Drug Amendments
these compounds were determined to the Federal Food, Drug, and Cosmetic Act that
based on data available to the FDA. drug products be approved for marketing based on a
demonstration of effectiveness as well as safety.
Potential Internal Standards (IS) are
2 Amidon, G. L., H. Lennernas, V. P. Shah, and J. R.
also identified. Crison, “A Theoretical Basis For a Biopharmaceutic
Tablet-A
Drug Classification: The Correlation of In Vitro Drug
Drug Product Permeability Product Dissolution and In Vivo Bioavailability,”
Class Pharmaceutical Research, 12: 413-420 (1995).
3 The United States Pharmacopeia, 23, January
Ketoprofen High 1995. United States Pharmacopeial Convention, Inc.
Rockville, Maryland.
DRAFT GUIDELINE
Naproxen High 4. Guidance for Industry, Dissolution Testing of
Immediate Release Solid Oral Dosage Forms, FDA,
Verapamil High CDER, August 1997.
5. Criteria for identifying a narrow therapeutic index
Carbamazepine (Narrow High drug are currently being developed by FDA.
Therapeutic Range Drug) 6 Guidance for industry, Immediate Release Solid
Oral Dosage Forms: Scale-Up and Post-Approval 7
Propranolol High Changes, November 1995.
7. International Journal of Generic Drugs, Vol. 02-No
02. 1998
Metoprolol High 8. Skelly, J. P., G. L. Amidon, W. H. Barr, L. Z.
[Potential IS Candidate] Benet, J. E. Carter, J. R. Robinson, V. P. Shah, and
A. Yacobi, 1990, "In Vitro and In Vivo Testing and
Theophyllin - Narrow High Correlation for Oral Controlled/Modified-Release
Therapeutic Range Drug Dosage Forms," Pharmaceutical Research, 7:975-
982.
9 FDA Narrow Therapeutic Range Drug List
Caffeine High
Inhalation Aerosol
Isoetharine Mesylate Inhalation Aerosol
Transdermal Patches
Clonidine Patch
HIGHLY SOLUBLE
ACTIVES - KETOROLAC
EVALUATING POTENTIAL DISSOLUTION
DIFFERENCES BETWEEN THE
Drug substance, Powder Blend & Compressed Tablets
DISSOLUTION PARAMETERS IN
VERY SOLUBLE ACTIVES
ESTABLISHING A CORRELATION BETWEEN
Drug substance, Powder Blend & Compressed Tablets
Table 1
DRUG SUBSTANCE - DISSOLUTION PROFILE
Percentage assay ketorolac tromethamine dissolved in:-
Method SI-00-00 Edition #00-00 RPM 50 Paddle 600mL Water
Median
10 min 20 min 30 min 45 min
Particle
size
13 85.0 ± 7.1 92.0 ± 3.8 96.0 ± 2.1 98.5 ± 1.1
23 84.0 ± 5.5 88.0 ± 4.2 91.0 ± 3.1 93.0 ± 2.1
55 87.0 ± 7.2 93.0 ± 3.4 96.0± 2.5 98.0 ± 0.9
Solubility at 25oC in water (pH6.8) = 0.55g/mL
Intrinsic Dissolution Rate at 25oC in HCl (pH2.0)= 16-18mg/min/mL
Ketorolac tromethamine has five polymorphic forms [XRD/DSC]; crystal forms I - IV and one
amorphous form. Crystal forms I, II and III have very similar crystal lattice energies, melting
points (162oC), solubilities and intrinsic dissolution rates.
References - 1. Berge, SM., Bighly, L.D., & Monkhouse, D.S., J. Pharm Sci. 66 (1977) p1-192.
Current Conceps in Pharmaceutical Sciences Dosage form Design and Bioavailability, Swarbrick
Lea & Febiger (1973, 18)
Food-Effects
IN BA-BE Studies
FOOD and FED STUDIES
IMPACT OF 'FOOD EFFECTS" IN BA-BE STUDIES
NDAs for MR Products: For ANDAs, the lot and strength tested
in the pivotal BE fasted study should be
Information on food effects should be
tested in the food-effect BE study.
obtained for the to-be-marketed When multiple strengths of MR drug
formulations of all NDAs for MR products are intended for marketing
products. and the food-effect BA or BE study is
Additional studies during MR performed on one of these strengths,
formulation development can be invitro dissolution testing should be
undertaken by sponsors who wish to conducted for all other strengths in
distinguish between food effects due to three different pH media.
drug substance versus effects due to Similarity of dissolution should be
formulation and/or processing factors. established in accordance with the f2
Major postapproval changes in test specified in SUPAC guidance
formulation and/or processing leading documents. Lack of similarity of
to a recommendation for a fasted BE dissolution could indicate that
study (see SUPAC-MR) should be additional food-effect studies should be
accompanied by a food-effect BE study. performed using other strengths.
Sponsors should provide justification ANDAs CR/DR/MR need
for not conducting this postapproval
study. 12 point Dissolution
MR/CR/DR ANDAs Similarity Testing
Require Fasted [D]. TEST MEAL
and Fed Studies The primary food-effect BA and BE
study should be conducted under
ANDAs for MR Products: conditions expected to provide maximal
All ANDAs for MR formulations should perturbation due to presence of food in
establish, in addition to BE under fasted the GI tract.
conditions, BE to the RLD formulation A high fat (approximately 50% of total
under fed conditions. caloric content of the meal), high calorie
Major postapproval changes in (approximately 1000 calories) breakfast is
formulation and/or processing involving therefore recommended as a test meal for
a fasted BE study should be food-effect BA and BE studies.
accompanied by a food-effect BE study. A representative example is 2 eggs fried in
Sponsors should provide justification for butter, 2 strips of bacon, 2 slices of toast
not conducting this postapproval study. with butter, 4 ounces of hash brown
Strength: potatoes, 8 ounces of whole milk (i.e.,
approximately 150 protein calories, 250
Generally, the highest strength of a carbohydrate calories, 500-600 fat
product should be tested in food-effect calories).
BA and BE studies. In some cases,
clinical safety concerns could warrant
use of lower strengths of the dosage Alternative meals with equivalent
form. nutritional content can be used. Details
of the meal should be recorded prior to
Highest Dosage Strength the study and provided in the study
is normally chosen report. The sponsor should provide a
scientific rationale, if the selected meal
For The Food-Effect BE is not high in calories and fat.
products
Fed treatments:
Following an overnight fast of at least Individual subject parameters, as well
10 hours, subjects should be served the as summary statistics (e.g., group
test meal and ingest this meal within 30 averages, standard deviations,
minutes. The drug product should be coefficients of variation, 90%
administered with 180 ml (6 fl oz) of confidence intervals [CI]) should be
water immediately (within 5 minutes) reported.
after completion of the meal.
In food-effect BA studies, the fasted
No food should be allowed for at least treatment serves as the reference. In
4 hours post-dose. Water can be
food-effect BE studies, the RLD product
allowed ad libitum after 2 hours.
Subjects should be served scheduled administered under fed conditions
standardized meals throughout the serves as the reference.
remaining study period.
The results for food-effect BA studies
[F]. SAMPLE COLLECTION will be evaluated according to the
For both treatment periods, timed following options:
biological fluid samples should be In BE Food Effect
collected from the subjects to permit
characterization of the complete plasma
the RLD Fed Study
concentration-time profile for drug is the reference
and/or metabolites. Food Effect Absent: Absence of a
food effect will be concluded when the
For Delayed Release 90% CI for the ratio of means
(Enteric Coated) Adjust (population geometric means based on
log-transformed data) of fed and fasted
blood sampling times treatments fall within 80%-125% for
Caution should be used when studying AUC and 70%-143% for Cmax
MR dosage forms (e.g., enteric coated Food Effect Documented: A food
products) where coadministration with effect will be concluded when the 90%
food can delay in vivo drug release. CI for the ratio of means (population
SIDE-BY-SIDE COMPARISON
BA & BE FOOD EFFECT STUDIES
Fasted
& Food
NDA ANDA
conducted
STUDIES STUDIES
on SAME
B/E Batch
ANALYTICAL VALIDATION
GUIDANCE FOR INDUSTRY
Bioanalytical Method
Validation for Biostudies
‘…designed for all biostudies and applies specifically to
generic comparative bioequivalency studies…
collection and the date of last sample The stability of both the drug and the
analysis. internal standard should be evaluated
Long-term stability should be in validation samples under these
determined by storing at least three conditions by determining
aliquots of each of the low and high concentrations on the basis of original
concentrations under the same calibration standards.
conditions as the study samples. Although the traditional approach of
A suggested storage temperature for comparing analytical results for stored
the majority of drugs and metabolites in samples with those for freshly prepared
a biological matrix is -20 C, but lower samples has been referred to in this
temperatures (e.g., -70 C) may be guidance, other statistical approaches
recommended to prevent degradation based on confidence limits are also
problems observed at higher available for the development of SOPs
temperatures. for evaluation of an analyte’s stability in
The volume of samples should be a biological matrix - (Timm 1985).
sufficient for analysis on three
occasions. SOPs to describe
The concentrations of all the stability Statistical methodology
samples should be compared to the
mean of back calculated values for the and rules used
standards at the appropriate
concentrations from the first day of
W hatever approach is used, the SOPs
should clearly describe the statistical
long-term stability testing (Buick 1990).
method and rules employed. Additional
4. Stock Solution Stability validation may include investigation of
The stability of stock solutions of drug samples from dosed subjects.
and the internal standard should be F. Acceptance Criteria
evaluated at room temperature for at
An analytical method is considered fully
least 6 hours. validated when it meets the following
The stability samples should then be criteria:
refrigerated or frozen for 7 to 14 days Precision: The between-batch CVs for
or other relevant period. low, medium, and high concentrations
After completion of the desired storage should be ≤15%, and ≤20% for the
time, the stability should be tested by LOQ QC, using a minimum of three
comparing the instrument response batches.
with that of freshly prepared solutions Accuracy: The between-batch mean
(Buick 1990). value should be within ±15% of the
nominal value at low, medium, and high
5. Autosampler Stability
QC concentrations and should not
The stability of processed samples in deviate by more than ±20% at the LOQ.
the auto-sampler should be determined
at the auto-sampler temperature that Sensitivity: The lowest standard
will be used during analysis, which is should be accepted as the limit of
usually room temperature, but may quantitation of the method if the
sometimes be a lower temperature between-batch CV at the LOQ QC is
(e.g., when a refrigerated auto-sampler ≤20%.
Specificity: The responses of
is used). Stability should be assessed
interfering peaks at the retention time
over the anticipated run time for the of the analyte should be less than 20%
batch size to be used in studies.
of the response of an LOQ standard.
PREPARATION
REFERENCES This guidance has been prepared by the
Biopharmaceutics Coordinating Committee and
Ayers, G., D. Burnett, A. Griffiths, and A.
the Clinical 1 Pharmacology Section of the
Richens, “Quality Control of Drug Assay,” Clin
Medical Policy Coordinating Committee in the
Pharmacokinetics 1981; 6:106-117.
Center for Drug Evaluation and Research
Brooks, M.A. and R.E. Weifeld, “A Validation
(CDER) at the Food and Drug Administration.
Process for Data from the Analysis of Drugs in
Biological Fluids,” Drug Development and
Industrial Pharmacy 1985; 11: 1703-1728. CURRENT AGENCY THINKING
Buick, A.R., M.V. Doig, S.C. Jeal, G.S. Land, This guidance document represents the
and R.D. McDowall, “Method Validation in the Agency’s current thinking on validation of
Bioanalytical Laboratory,” Journal of analytical methods for human studies based on
Pharmaceutical and Biomedical Analysis 1990; drug or metabolite assay in a biological matrix.
8:629-637. ALTERNATIVE APPROACHES
Karnes, S.T., G. Shiu, and V.P. Shah, It does not create or confer any rights for or on
“Validation of Bioanalytical Methods,” any person and does not operate to bind FDA
Pharmaceutical Research 1991; 8:421-426. or the public.
Mehta, A.C., “The Validation Criteria for An alternative approach may be used if such
Analytical Methods used in Pharmacy Practice approach satisfies the requirements of the
Research,” J Clin Pharm Ther 1989; 14:465- applicable statute, regulations, or both.
473.
Pachla, L.A., D.S. Wright, and D.L. Reynolds,
“Bioanalytical Considerations for DRAFT RELEASE DATE
Pharmacokinetic and Biopharmaceutic RELEASE DATE OF THIS DRAFT GUIDANCE
Studies,” J Clin Pharmacol 1986; 26:332-335. December 1998
Shah, V.P., K.K.Midha, S.V.Dighe, et al.,
Analytical Methods Validation: Bioavailability, U.S. Department of Health and Human Services
Bioequivalence and Pharmacokinetic Studies Food and Drug Administration Center for Drug
(Conference report). Pharmaceutical Research Evaluation and Research (CDER) Dec. 1998 BP # [ ]
1992; 9:588-592. DOCUMENT CODE j:\!guidance\2578dft.wpd 12/14/98
ØC H E C K L I S T ×
CL # P-HB01-03-1Y2K
1. Has theGC / HPLC analytical method been fully validated in the oYes qNo
appropriate biological matrix (blood, serum, plasma, or urine) prior to the
Start of the Study
2. Does the pre-study written validation protocol include all the listed six oYes qNo
[6] parameters as well as stability of the relevant matrix?
3. Has the firm a complete set of SOPs exist covering record keeping, oYes qNo
security and chain of sample custody (accountability systems) to ensure
the integrity of test articles from pre-sampling procedures to final report?
4. Can the analyte in the test samples be evaluated in an interference- oYes qNo
free biological matrix for each sample (i.e. for each individual subject.)?
5. Has the validated analytical method met the control percentages allow o Yes qNo
for Accuracy Precision Sensitivity Specificity and Stability criteria?
6. Has a standard calibration curve covering the expected unknown oYes qNo
sample concentration range in addition to a calibrator sample at LOQ
been established?
7. Have all subject samples been analysed in a single continuous test oYes qNo
run? (one sitting?)
8. Are QC samples analyzed concomitantly with the test samples at oYes qNo
intervals to test accuracy and precision of the VALIDATED method?
9. Do the test samples meet the criteria spread of 'Near the LOQ' (i.e., oYes qNo
#3 x LOQ), 'midrange', and 'High end range?
10. Are Re-assay SOPS specifying exact retest conditions fully in place? oYes qNo
11. Is there a set of written SOPs controlling The Protocol - The Test oYes qNo
Method, - The method Validation and the final Assay Validation Report?
12. Is a written protocol available for the analytical method PRIOR to oYes qNo
validation
13. Has the analytical method been fully validated, documented with the oYes qNo
presentation of a final Assay Validation Report?
14. Have the Lab books been corrected signed and audited? oYes qNo
15. Has all Study Documentation is subject to a Comprehensive Audit? oYes qNo
Page: 1 of 8.
PURPOSE:
The purpose of this Standard Operating Procedure is to perform the following
performance verification & calibration test procedures on the dissolution apparatus
In order to ensure the functioning of apparatus at parameters prescribed by USP/BP
Pharmacopoeias and by the September 1997 Approved Guidance to Industry:-
Performance Verification is carried out by:-
n Routine calibration & checking
n Eccentricity of shafts
n Apparatus suitability
n Calibration & checking of apparatus
RESPONSIBILITY:
1. Routine Checking & Calibration Procedure
Each analyst shall verify the apparatus suitability prior to use.
2. Eccentricity of Shafts
The analyst in charge of the apparatus shall check the eccentricity of the shafts.
3. Apparatus Suitability
The analyst in charge of the apparatus shall verify that the apparatus is suitable for
performing dissolution tests.
4. Calibration & Checking of Apparatus
The appointed laboratory technician shall check the correct functioning of the
apparatus.
FREQUENCY:
1. Routine Calibration
Immediately prior to each dissolution testing procedure.
2. Eccentricity of Shafts
Once a month or when shafts are changed.
3. Apparatus Suitability
Every six months and after any maintenance procedure.
4. Calibration & Checking of Apparatus
Annually and after any maintenance procedure.
Page: 2 of 8.
PROCEDURE:
Routine Calibration (See Form 01/9Y).
1. Vessels:-
Inspect each vessel for any defects (cracks, etc.), and ensure that the numbered
vessels are in the correct positions.
After the test has been performed, make sure that each vessel is thoroughly cleaned
and safely stored.
2. Centering:-
Place vessels through their holes in the bath cover. Center each vessel using a
suitable centering unit with respect to the vertical axis of its shaft.
4. Medium:-
Dissolution Media should be vacuum filtered if necessary.
Using a graduated cylinder, place the specified amount of media pre-warmed to
approximately 37°C, or media at room temperature, into each vessel. Allow the
medium to assume the temperature of the water bath (37° ±0.5°C ). Observe the
stirrer and the walls of the vessel: No air bubbles should be noted.
5. Temperature:-
Prior to starting the test, check the temperature of the Dissolution Medium in each
vessel (37 ±0.5°C). Use a calibrated thermometer.
Page: 3 of 8.
8. Covers:-
Place the appropriate cover over each vessel.
9. Visual Observation:-
Observe the remaining contents of the baskets/vessels after dissolution. Record
remarks on attachment Form "Routine checking and calibration" (See Form 01/9Y).
Page: 4 of 8.
Procedure
Place the stated volume of Dissolution Medium in the vessels of the apparatus,
assemble the apparatus, equilibrate the Dissolution Medium to 37.0 ± 0.5°C and
remove the thermometer.
Place 1 tablet in each vessel, or in each dry basket, taking care to exclude air
bubbles from the surface of the tablets, and immediately operate the apparatus at
the stated rate.
After the stated time passes, withdraw a specimen of solution from a zone midway
between the surface of the Dissolution Medium and the top of the rotating paddle or
basket, not less than 1 cm from the vessel wall.
Filter, using a 0.45µ membrane filter HAWP (or similar), discarding the first few mL
of the filtrate.
Standard Solution
Transfer about 20 mg of Salicylic Acid USP Reference Standard, accurately
weighed, to a 200-mL volumetric flask. Add about 2 mL of Ethanol and sonicate for
about 2 minutes until dissolved, allow the solution to cool to room temperature.
Dilute with Dissolution Medium to volume and mix. Filter using a 0.45µ membrane
filter HAWP (or similar) discarding the first few mL of the filtrate.
Pipette 5 mL of filtered Standard solution into a 25-mL volumetric flask, dilute with
Dissolution Medium to volume and mix, (or dilute 1000µL of the filtered Standard
solution with 4000µL Dissolution Medium by the aid of a dilutor [Hamilton] ).
Determine the amount of salicylic acid dissolved from ultraviolet absorbances at the
wavelength of maximum absorbance at about 296 nm of Sample solution in
comparison with the Standard solution, using the Dissolution Medium as the blank.
Page: 5 of 8.
Au x Wst x 5 x 900 x 25 =
As 200 25 300 5
Au
x Wst x 1.5 = % of the labeled amount of SALICYLIC ACID dissolved per tablet
As
4.3.2.1 Apparatus:
Apparatus I (Basket) (See Form 05/9Y).
Apparatus II (Paddle) (See Form 06/9Y).
Page: 6 of 8.
After the stated time passes, withdraw a specimen of solution from the zone midway
between the surface of the Dissolution Medium and the top of the rotating paddle or
basket, not less than 1 cm from the vessel wall.
Filter, using a 0.45µ membrane filter HAWP (or similar), discarding the first few mL
of the filtrate.
Ü Apparatus I, 50 rpm
Use the filtered solution with no further dilution.
Ü Apparatus I, 100 rpm;
Ü Apparatus II, 50 and 100 rpm
Pipet 4 mL of the filtered solution into a 10 mL volumetric flask, dilute to volume with
purified water and mix (or dilute 2000 µL of the filtered solution with 3000 µL purified
water by dilutor).
Standard Solution
Transfer about 10 mg of Prednisone USP Reference Standard, accurately weighed,
to a 50 mL volumetric flask. Add about 5 mL of Ethanol and sonicate for about 2
minutes until dissolved, allow the solution to cool to room temperature.
Dilute with purified water to volume and mix. Filter using a 0.45µ membrane filter
HAWP (or similar) discarding the first few mL of the filtrate.
Pipette 5 mL of Standard solution into a 100 mL volumetric flask, dilute with purified
water to volume and mix (or dilute 250 ml of the filtered standard solution with a
4750 ml Dissolution Medium using a dilutor).
Determine the amount of prednisone dissolved from ultraviolet absorbances at the
wavelength of maximum absorbance at approximately 242 nm of Sample Solution in
comparison with the Standard Solution, using Purified Water USP as the blank.
Page 7 of 8.
LIMITS/LIMITATIONS
Routine Calibration Specification Limits
[1] Revolutions per minute → Specified rpm ±4%
[2] Temperature - Celsius → 37° ±0.5°C
[3] Paddle or basket depth → 25 ± 2 mm
[4] Eccentricity of shafts: → 2.0 mm [basket]
→ 1.0 mm [paddle].
[5] Apparatus Suitability
The test limits are stated in the certificate for the USP dissolution calibrator (non-
disintegrating type and disintegrating type).
CORRECTIVE ACTION:
[1] Routine Calibration
If, during verification, the rpm is not within the specified range, immediately contact
the technician.
[2] Eccentricity of Shafts
If during verification the eccentricity of the shafts is not within the specified range,
repeat the verification with a new paddle or basket shafts.
r successful verification - discarded old shaft.
r unsuccessful verification - do not use and contact the technician immediately.
Page 8 of 8.
[3] Apparatus Suitability
If the individual calculated values at each indicated speed are not within the
specified ranges as shown in table I or II in the certificate for the USP dissolution
calibrator, the following system should be verified:
þ Routine calibration
þ Eccentricity of shafts
þ Performance verification of analytical balance (Monthly check according to SOP)
þ Dissolution medium
[4] Calibrator Suitability
n If all of the above are found to be in correct working order, open another USP
dissolution calibrator (Salicylic Acid Tablets, 300 mg., or Prednisone Tablets, 50
mg.) and repeat the test.
n If the test is within the specified range, discard the old calibrator.
n If not, shut down the apparatus and immediately contact the lab technician. Place
a "Temporarily Out of Order" sign on the apparatus.
DOCUMENTATION:
Form 01 - "Routine Checking and Calibration".
Form 02 - "Eccentricity of Shafts".
Form 03 - "Apparatus Suitability - Salicylic Acid" (Basket Method)
Form 04 - "Apparatus Suitability - Salicylic Acid" (Paddle Method)
Form 05 - "Apparatus Suitability - Prednisone Tablets" (Basket Method)
Form 06 - "Apparatus Suitability - Prednisone Tablets" (Paddle Method)
PERFORMANCE VERIFICATION OF
DISSOLUTION APPARATUS
ECCENTRICITY OF SHAFTS
Dissolution Apparatus ______________________ No ______________
Shaft Paddle Limit Basket Limit
Number
1
2
3 1.0 mm 2.0 mm
4
5
6
Form: 02/ 9Y
REMARKS________________________________________________________________
__________________________________________________________________________
__________________________________________________________________________
PERFORMANCE VERIFICATION OF
DISSOLUTION APPARATUS
APPARATUS SUITABILITY
SALICYLIC ACID STANDARD
[Basket Method]
USP Non-disintegrating Type-SALICYLIC ACID Tablets, 300 mg. Lot No. ____________
USP SALICYLIC ACID Reference Standard Lot No.: ____________
BASKET METHOD
q RPM: 50 q RPM: 100
Limits: ___________ Limits: ____________
q Approved: ___________________
q Rejected: ___________________
REMARKS:
__________________________________________________________________________
__________________________________________________________________________
PERFORMANCE VERIFICATION OF
DISSOLUTION APPARATUS
APPARATUS SUITABILITY
SALICYLIC ACID STANDARD
(Paddle Method)
USP Salicylic Acid Reference Standard Lot No.: _________
USP Non-disintegrating Type-SALICYLIC ACID Tablets, 300 mg. Lot No. ___________
USP SALICYLIC ACID Reference Standard Lot No.: ___________
PADDLE METHOD
q RPM: 50 q RPM: 100
Limits: ___________ Limits: ____________
q Approved: ___________________
q Rejected: ___________________
Remarks:
________________________________________________________________________
PERFORMANCE VERIFICATION OF
DISSOLUTION APPARATUS
APPARATUS SUITABILITY
PREDNISONE STANDARD TABLETS
[Basket Method]
BASKET METHOD
q RPM: 50 q RPM: 100
Limits: ___________ Limits: ____________
q Approved: ___________________
q Rejected: ___________________
REMARKS:
___________________________________________________________________________
___________________________________________________________________________
OPERATOR: ______________ SUPERVISOR: __________________ Date: __________
PERFORMANCE VERIFICATION OF
DISSOLUTION APPARATUS
APPARATUS SUITABILITY
PREDNISONE STANDARD TABLETS
(Paddle Method)
PADDLE METHOD
q RPM: 50 q RPM: 100
Limits: ___________ Limits: ____________
q Approved: ___________________
q Rejected: ___________________
Remarks:
___________________________________________________________________________
___________________________________________________________________
False Positives
EXAMINED
Chow, Pitt
Similarity Testing and
Others
Similarity (f2)
Pitfalls
'Points to Consider'
A REVIEW DISSOLUTION SIMILARITY EVALUATION
IMPACT OF 'SIMILARITY TESTING" IN DISSOLUTION PROFILES
If the variance is large, then the The sensitivity of split-plot and two-way
probability of declaring a difference with ANOVA are decreasing functions of
Chow’s method is even larger than other variation.
methods no matter what change exists in Fda' and Chow's
the initial amount or rate of dissolution. methods are more
Generally the FDA's relaxed than others
SIMILARITY Equation is less In general, the FDA and Chow methods
discriminatory are more relaxed than the others.
than Chow Although FDA’s similarity factor is easy to
manipulate and results do not change if
In cases where dissolution profiles are the role of test and reference interchange,
quadratic or can be approximated by a it, however, lacks scientific justification
quadratic model, if the amount dissolved and has the following disadvantages:
is the same for two drug products at two [1]. It is not based on an hypothesis
boundaries but different in the dissolving testing procedure, therefore, there is no
rate, neither the FDA method nor the measurement of the associated error
Chow method are sensitive to the (Type I or II errors)
differences and have zero probability of [2]. It is a function of the pair difference
declaring a difference of less than 15% between two drug products. If the time
(FDA) or 20% (Chow). points selected for two drug products are
When the variation is large, Chow has a different, the method could not be
large probability of declaring a difference applied
even when two profiles are identical (eg., [3]. The measurement is an average of
probability = 50% for s =6) but FDA stays differences observed. After the asymptote
the same. is achieved, the differences could be fairly
The other methods detect the difference small.
right away (100% for even a slight In the guideline, no time point selection is
difference). If two profiles are the same specified. If the analyst selects a large
before 45 minutes and both have Q0.75 = number of time points after the asymptote
75% but differ afterwards, the same is achieved, then the average difference
pattern as the earlier case is observed could be small.
with FDA and Chow and they are even In consequence, f2 will tend to have a
less sensitive to the difference. value between 50 and 100 which results
If two profiles are different except for the in similarity although two profiles could be
origin in the study range where the very different.
difference between R and T are within 5% Too many sampling
at Q0.75 and Q2, FDA and Chow both have
zero probability of detecting the difference points after asymptote
when the variation is small. The same can distort the
pattern is observed for Chow for its SIMILARITY result
sensitivity to the variation (eg., 50% for s
=6 and 75% for s =8 even when two Chow’s method has an advantage in that
profiles are identical). it defines the equivalence limits for
For the second order model, ANCOVA similarity based on Q value when
specified in the appropriate USP
has a small probability of declaring the
monograph method.
difference, which may due to its linear
relationship assumption. It also takes into account that the
dissolution series observed are in fact
dependent and the dependency is a For example, the interval derived from the
decreasing function of time, as opposed auto correlation2 model for the data in
to Gill’s method where the relationship is Tsong and Hammerstrom5 is within two
assumed to be constant over time. tenth points of the auto correlation model1
It however, has the following (i.e. 86.7% and 112.9%)
disadvantages and weaknesses:
Gill's method uses
Chow' s Method Time as the Variable -
Depends on (Q)
a Limiting Disadvantage
excludes T0 and requires
identical procedures For Gill’s method, the advantage is that it
reflects the fact that correlation between
CHOWS DISADVANTAGES
the amount dissolved for the same
[1] When time T0 is included, Chow’s
product exists.
method will declare dissimilarity for almost
100% regardless of the variation. It can be This assumption, however, seems
explained that Chow’s test statistic is, in oversimplified since it is assumed to be
fact, a ratio of two random deviates. If the constant. In addition, due to the algorithm
denominator is close to 0, then R is large. used in hypothesis testing, a Type I error
Therefore, the probability of concluding obtained is twice as high as
dissimilarity is also large. it should be since dissimilarity could be
[2]. If Q is not specified, the method is concluded in two cases, that is, if the
difficult to implement in actual practice, treatment by time interaction exists or if
[3] The method is based on relative the difference between treatments is
dissolution that requires the same number found in the reduced model.
of time points and the same number of Moreover, using time as a class variable
locations (e.g., baskets) for the Test & in the analysis dramatically reduces the
RLD in the evaluation. degrees of freedom available and thus a
If either one is different, the method could disadvantage of the method.
not be applied,
[4]. The response is based on the ratio of ANCOVA Estimates
test over reference. - If the role of two
drug products is interchanged in the Rate & Similarity
computation, the resulting confidence
interval will vary. In consequence, it might
but
lead to a different conclusion, needs Special Conditions
[5]. It is very sensitive to the variation. Ithas been observed that ANCOVA and
If the variation is large, the probability of two-way ANOVA are often used in
declaring similarity is large even when the comparing dissolution profiles.
underlying distribution of two profiles is
identical. ANCOVA especially is preferred by most
since it can estimate the dissolution rate
Chow’s method may be extended into the of drug products other than testing the
auto correlation2 model, which takes into similarity of the profiles.
account the correlation of more than one
time point. ANCOVA
The results achieved are very similar to
the auto correlation model1 with a slightly
needs at least 5-6
wider confidence interval. time points
It should be noted that the method is not 7. Pena Romero A, et. al. Water uptake and force
appropriate if the dissolution profiles are development in an optimized prolonged release
not linear or if only a few time points are formulation. Int J Pharm. 1991;73:239–248.
8. Langenbucher F. Linearization of dissolution
tested. rate curves by the Weilbull distribution. J Pharm
Choose the Test Pharmacol. 1972;24:979–981.
9. Kervinen L, Yliruusi J. Modelling S-Shaped
Procedure for the dissolution curves. Int. J. Pharm. 1993 ;92:115 –
122.
Required
Sons; 1991.
11. Mauger JW, et al. On the analysis of
dissolution data. Drug Dev Ind Pharm. 1986:12(7)
Moreover, the correlation nature of the :969–992.
12. Gill JL. Repeated measurement: split-plot
dissolution series within the same product trend analysis versus analysis of first differences.
violates the underlying independent Biometrics. 1988; 44:289–297.
assumptions required for applying either 13. Tsong Y. Statistical assessment of mean
of the methods. differences between two dissolution data sets.
Presented at the DIA Dissolution Workshop,
The validity of the results is, therefore, Rockville, Maryland, 1995.
questionable. 14. Box GEP, et al. Time Series Analysis,
Forecasting and Control. 3rd ed. Englewood Cliffs,
NJ: Prentice Hall; 1994.
15. Shah VP, Lesko LJ. Current challenges and
future regulatory directions in in vitro dissolution.
REFERENCES Drug Info J. 1995;885–891.
16. Leeson LJ. In vitro/in vivo correlations. Drug
1. FDA. Guidance for industry: immediate release Info J. 1995; 903–915.
solid oral dosage forms scale-up and post- 17. Chow SC, Ki FYC. Statistical comparison
approval changes: chemistry, manufacturing and between dissolution profiles of drug products. J
controls, in vitro dissolution testing, and in vivo Biopharma Stat. 1997;7:241–258
bioequivalence documentation. Rockville, MD: 18. Chow SC, Liu JP. Current issues in
Food and Drug Administration; November 15, bioequivalence trials. Drug Inf J. 1995;29:795–
1995. 804.
2. Chow SC. Statistical comparison between Chow SC, Liu JP, Ma MC Statistical evaluation of
dissolution profiles of drug products. Presented at similarity factor f2 for assessment of similarity
Department of Health, Executive Yuan, Taipei, between dissolution profiles Drug Inf J.
Taiwan, 1995. 1997;31:1255–1271
3. Chow SC, Liu JP. Statistical Design and 19.Metzler CM. Bioavailability: A problem in
Analysis in Pharmaceutical Science. New York: equivalence. Biometrics. 1974;30:309–317.
Marcel Dekker; 1995. 20. Chow SC, Liu JP. Design and Analysis of
4. USP/NF. The United States Pharmacopoeia Bioavailability and Bioequivalence Studies. New
XXIII and the National Formulary. Rockville, York:
Maryland:The United States Pharmacopeial 21. Leeson LJ. In vitro/vivo correlation. Drug Inf. J.
Convention; 1990. 1995;29:903–915.
5. Tsong Y, Hammerstrom T. Statistical issues in 22. FDA. Guidance on Statistical Procedures for
drug quality control based on dissolution testing. Bioequivalence Studies Using a Standard Two-
Proceedings of Biopharmaceutical section of Treatment Crossover Design. Rockville, Maryland:
American Statistical Association, 1994;295–300. Division of Bioequivalence Office of Generic
Tsong Y. Statistical assessment of mean Drugs, Food and Drug Administration.
differences,. between two dissolution data sets. 23. Moore JW, Flanner HH. Mathematical
Presented at the 1995 Drug Information comparison of curves with an emphasis on
Association Dissolution Workshop, Rockville, dissolution profiles. 1992 Pharma Technol.
Maryland. 6. Dawoodbhai S, et. al. Optimization of 1996;20:64–74
tablet formulation containing talc. Drug Dev Ind 24. Serfling RJ. Approximation Theorems of
Pharm. 1991;17: 1343–1371. Mathematical Marcel Dekker, Inc.; 1992.
TTD
Technical Transfer
Documentation-Pharmaceutical
TTD
Technical Transfer
Documentation
‘the time has come the researcher said to speak of many things
- of pivotal and protocols and to validated cleanings...’
ØC H E C K L I S T ×
CL # P-HGD-03-01Y2K
1. The buying department has purchasing specifications for the qYes qNo
procurement of approved actives, excipients and container-closure
systems ?
2. Printing specification and QA approved artwork for product labels, qYes qNo
cartons, package inserts and advertising claims are approved ?
3. The manufacturing formula and master processing instructions for each qYes qNo
commercial batch sizes are approved ?
4. Cleaning validation protocol specific to the active material is complete ? qYes qNo
5. The validation protocol for the first three consecutive batches is qYes qNo
approved?
6. All new manufacturing equipment has been fully qualified (IQOQ)? qYes qNo
7. The metrology department has calibrated all equipment recording qYes qNo
units?
8. Plant QC laboratory has evaluated the transferred analytical methods qYes qNo
for system suitability and robustness (ruggedness) ?
9. Microbiological laboratory has all methods and specifications? qYes qNo
10. Physical QC lab has all container-closure methods and qYes qNo
specifications?
11. All product specific SOPs have been distributed and signed as ‘read qYes qNo
and understood’ by the production operators and supervisors?
12. Stability unit has the ANDA commitment stability protocol (real-time qYes qNo
study) and the validation stability protocol for first three consecutive
batches?
13. All vendor DMF deficiencies (and GMP concerns) have been qYes qNo
corrected?
14. The CMC file is compiled in full and signed-off ? qYes qNo
15. The Product ‘Development Report’ is complete and signed-off ? qYes qNo
STANDARD OPERATING
PROCEDURES
CL # P-HGD-03-01Y2K Page 1 of 1.
4
[End of Document]
STANDARD OPERATING
PROCEDURES
SOP # TTD-01-06Y2K
2. RESPONSIBILITY
The Head of Pharmaceutical Development together with the Responsible Researcher
for the Generic Drug Development Project.
3. FREQUENCY
Each ANDA product formula developed for the US market.
4. PROCEDURE or SCOPE
[4.1]. The responsible personnel for the product development will prepare the
pharmaceutical section of the technical transfer file (TTD) for process and data
information transfer to the commercial manufacturing site facility.
[4.2]. The Pharmaceutical TTD file will contain all necessary pharmaceutical master
formula, manufacturing methods, validation criteria, product specifications, technical
data, reports, tabulations and summaries based on the pharmaceutical development
work pertaining to all strengths of the generic drug development from the pre-
formulation to process qualification stage.
This data is required for the manufacture and control the pivotal submission batch
and the three initial full size validation batches produced at the commercial
manufacturing site facility.
[4.3]. Each section of the TTD File is presented in a modular form for ease of
updating. Sections are numbered [A] to [K]. The outline of the TTD requirements is
presented in a standard operating procedure format.
[4.4]. The requirements of the Pharmaceutical Technical Transfer documentation will
be part of the Product Development SOP for the specific dosage form. This
documents is based on formulation development for oral tablet dosage forms.
[4.5]. An pharmaceutical TTD SOP will be prepared for each separate generic dosage
form under product development. 3
STANDARD OPERATING
PROCEDURES
SOP # TTD-01-06Y2K
STANDARD OPERATING
PROCEDURES
SOP # TTD-01-06Y2K
STANDARD OPERATING
SOP # A-00-01-Y2K
PROCEDURES
Page: 1 of 3
1. PURPOSE
The purpose of this Standard Operating Procedure is to establish the overall table
of contents for the preparation of the analytical part or section of the technical
transfer file for product information transfer to the selected commercial
manufacturing site facility. This SOP is specific for ANDA preparations of solid oral
dosage forms.
2. RESPONSIBILITY
The Head of Analytical Development together with the Responsible Researcher for
the Generic Drug Development Project.
3. FREQUENCY
Each ANDA product formula under development intended for the US market.
4. PROCEDURE or SCOPE
[4.1]. The responsible personnel for the product development will prepare the
analytical section of the technical transfer file (TTD) for method and data information
transfer to the commercial manufacturing site facility.
[4.2]. The Analytical TTD file will contain all necessary analytical methods, method
validations, product specifications, technical data, reports, tabulations and
summaries based on the analytical development work pertaining to the generic drug
development from the pre-formulation to process qualification stage.
This data is required for testing and analyzing the pivotal submission batch and the
three initial full size validation batches produced at the commercial manufacturing
site facility.
[4.3]. Each section of the TTD File is presented in a modular form for ease of
updating. Sections are numbered [A] to [G]. The outline of the TTD requirements is
presented in the standard operating procedure format.
[4.4]. The requirements of the Analytical Technical Transfer documentation will be
part of the Product Development SOP for the specific dosage form. This documents
is based on a Q&Q formulation development for solid oral dosage forms.
[4.5]. An analytical TTD SOP will be prepared for each separate generic dosage form
under product development.
4
[End of Page]
STANDARD OPERATING
SOP # A-00-01-Y2K
PROCEDURES
STANDARD OPERATING
SOP # A-00-01-Y2K
PROCEDURES
Process Validation
‘…process validation needs to show that the data from the first three
commercial lots of each batch size are essentially similar and compare
well with product data of the biobatch…’
ØC H E C K L I S T ×
CL # PV-05-03-01Y2K
1 The manufacturing product and processing controls of the commercial lots are qYes qNo
substantially the same (equivalent) as those listed in the ANDA filing?
2. Before the validation process and validation batches are run, the firm has qYes qNo
conducted a thorough and comprehensive on-site review and audit of all
development and analytical raw data?
3. The firm has performed a side-by-side evaluation of the validation protocol and qYes qNo
the submitted ANDA file to ensure that the product and process specification and
control points are common in both documents?
4. QC conducts routine particle size and bulk density analysis on the active qYes qNo
material(s) present in the drug product (a particle size range is given)?
5. Particle size and bulk density QC specifications have been established for the qYes qNo
active material(s) present in the final product formula?
6. During the product development the finished product has been evaluated at qYes qNo
both ends of the particle size and bulk density ranges and suitable meet the
finished product specification?
7. Process controls on granulator-mixers (time, rpm and settings) are qYes qNo
documented in the full scale commercial process instructions?
8. Uniformity of Content is a critical processing parameter for the bulk qYes qNo
granulation material after the blending process and prior to compression?
9. Granulation samples taken during sampling operations - for the assay qYes qNo
determination - are equivalent to one dosage unit?
10. In the event that it is not possible to obtain the equivalent of one dosage qYes qNo
unit in granulation samples, the closest approximation should be sampled (a
function of sampling thief used)?
11. A tapered sampling thief may during sampling operations be unsatisfactory qYes qNo
for sampling the equivalent of one dosage unit uniformly (use uniform diameter
thieves)?
12. Uniformity of Content is evaluated after the blending process with and qYes qNo
without lubricants added ?
13. Manufacturing instructions clearly indicate corrective action taken where qYes qNo
older mixers or blenders (pony / ribbon) display ‘dead spots’ where no direct
mixing action can occur (around shaft / bearing area or discharge valves) ?
ØC H E C K L I S T ×
CL # PV-05-03-01Y2K
14. Dedicated FBD bags are used for drug products containing colored qYes qNo
or insoluble active drug substances?
15. All critical process parameter temperature and time controls qYes qNo
incorporate automatic recording equipment?.
16. The product batch number and date is routinely entered on the qYes qNo
automatically recording temperature and time control graphs?
17. Dissolution profiles are performed on 12 dosage units for each of qYes qNo
the three validation lots. Note: Six units are appropriate when
generating the Q release data.
18. Where validation batches are used for sale then routine Finished qYes qNo
Product Testing must be performed on composite sample representative
of the overall production run.
19 This sampling procedure is a routine procedure and independent qYes qNo
and separate to the validation sampling requirements?
20. Assay, Content Uniformity, and Dissolution testing are critical qYes qNo
parameters and results are compared side-by-side to the bioequivalent /
pivotal batch for equivalency?
21. Equivalency of the bioequivalent / pivotal batch with the validation qYes qNo
batch lots implies a variation < 5% from the filed Biobatch?
22. All results, including failing results (if any) have been fully qYes qNo
discussed and explained in the validation report?
23. The basis for concluding that the validation process is satisfactory, qYes qNo
particularly those batch lots with failing results has been fully evaluated
in the validation report?
24. Any out-of-specification product or process result during validation qYes qNo
will be fully investigated and an Investigation Report will be completed
and signed-off within 30 days ?
25. A separate ‘Validation Concluding Statement’ is included in the qYes qNo
validation report? (Place on the approval page).
26. The ‘validation concluding statement’ indicates that the three qYes qNo
production lots are equivalent in all aspects to the filed ANDA batch?
STANDARD OPERATING
PROCEDURES
CL # PV-05-03-01Y2K Page 1 of 1.
P- HBPD - 10-01Y2K Do’s and Don’ts when preparing for Validation Batches.
4
[End of Document]
Validation Batch
GUIDELINES
‘…The validation batch is proof positive that the commercial process really works…‘
T
hese Guidelines are suitable for
challenge every aspect of the
post pivotal commercial
manufacturing filling and packaging
validation of solid dosage forms.
process and written instructions and
The guidelines are supplemented by
then test that all written product
various guideline SOPs, flowcharts
specifications are within limits.
and process validation checklists.
NOTE:-The minimum number of
batches is three lots per strength.
The Guidelines are suitable for a:-
What documentation must I
1. Oral IR Tablet Dosage Form have?
2. Oral IR Capsule Dosage Form Documentation must be
3. Solid Oral MR/CR Dosage Complete i.e.
Form PRODUCTION
Full master formula and manufacturing
Prior to starting the Process instructions for each batch size and a
Validation, each validation procedure master validation SOP plus validation
requires the following FIVE key protocol with all supporting analytical
documentation packages. test documentation.
ANALYTICAL
a. General Validation Guideline The assay and dissolution test must be
b. Validation Master SOP fully validated at this stage and be
c. Validation Protocol (Specific) 'essentially similar' the pivotal or
d. Validation Checklist (Specific) demonstration batch.
e. Validation Flowchart (Specific) Where are the 3 validation lots
made?
At the end of the Process Validation a Full Commercial conditions
Process Validation Report is prepared. The three Validation batches are
manufactured in the plant’s
This Process Validation Report commercial facilities where the
contains the test results and marketing lots will be produced, using
conclusions of the Validation Protocol standard production raw materials
specific to the dosage form evaluated from the warehouse and production
personnel, as well as routine QA
f. Product Validation Report. procedures.
ØG u i d e l i n e s ×
CL # CVG-S04-Y2K
PROCESS VALIDATION
‘…well developed process validation batch lots with good in-process controls
and excellent documentation will ensure failure-free COMMERCIAL production…’
1. The Process validation batch is equal to 100% of the smallest, middle or qYes qNo
largest commercial batch size? Each size must be validate separately.
2. The active material source has been qualified as an ‘Approved Supplier’ ? qYes qNo
3. All non actives are routine production excipients and have been approved? qYes qNo
4. The container closure-system is the final chosen marketing pack? qYes qNo
5. The Master Product Formula Record has all authorization signatures ? qYes qNo
6. The Master Manufacturing Batch Instructions has all authorization qYes qNo
signatures?
7. The manufacturing flow chart (identifying all manufacturing equipment and qYes qNo
process parameters) is final with all authorization signatures?
8. In-process QC specifications and processing parameters are complete? qYes qNo
9. Standard packaging instruction (including sampling protocol) are complete? qYes qNo
10. Release Specifications (with narrower lower and upper limits) are complete? qYes qNo
11. Check Specifications (with maximum lower and upper limits) are complete? qYes qNo
12. Overall Finished Product Specifications are complete? qYes qNo
13. The process validation PROTOCOL is written and signed-off ? qYes qNo
14. The analytical assay methods and stability indicating assay are complete? qYes qNo
15. The process validation is a full commercial batch performed after the qYes qNo
PIVOTAL batch
16. The Content Uniformity PROTOCOL is prepared for evaluation during qYes qNo
process validation batch manufacture.
17. The Uniformity of Content PROTOCOL will be evaluated during the qYes qNo
process validation batch manufacture?
18. Hardness Vs Dissolution testing (target value and range extremes) have qYes qNo
been performed, so as to produce an acceptable dissolution profile at all harness
values
ØG u i d e l i n e s ×
CL # PVG-S04-Y2K
PROCESS VALIDATION
‘…process validation needs to show that the data from the first
three commercial lots of each batch size are essentially similar
and compare well with product data of the biobatch…’
19. All critical process parameter temperature and time controls qYes qNo
incorporate automatic recording equipment?.
20. The product batch number and date is routinely entered on the qYes qNo
automatically recording temperature and time control graphs?
21. Dissolution profiles are performed on 12 dosage units for each of qYes qNo
the three validation lots.
Note: Six units are appropriate when generating the QC release data.
22. Where validation batches are used for sale then routine Finished qYes qNo
Product Testing must be performed on composite sample representative
of the overall production run.
23. This sampling procedure is a routine procedure and independent qYes qNo
and separate to the validation sampling requirements?
24. Assay, Content Uniformity, and Dissolution testing are critical qYes qNo
parameters and results are compared side-by-side to the bioequivalent/
pivotal batch for equivalency?
25. Equivalency of the bioequivalent / pivotal batch with the validation qYes qNo
batch lots implies a variation < 5% from the filed Biobatch?
26. All results, including failing results (if any) have been fully qYes qNo
discussed and explained in the validation report?
27. The basis for concluding that the validation process is satisfactory, qYes qNo
particularly those batch lots with failing results has been fully evaluated
in the validation report?
28. Any out-of-specification product or process result during validation qYes qNo
will be fully investigated and an Investigation Report will be completed
and signed-off within 30 days ?
30. The ‘validation concluding statement’ indicates that the three qYes qNo
production lots are equivalent in all aspects to the filed ANDA batch?
ØG u i d e l i n e s ×
CL # PVG-S03-Y2K
PROCESS VALIDATION
Commercial LOT Requirements
SOPs & Protocols - (Master Documentation Preparation)
1. Master Validation SOP is prepared and approved qYes qNo
2. VALIDATION PROTOCOL prepared and approved qYes qNo
3. CONTENT UNIFORMITY PROTOCOL prepared and approved qYes qNo
4. HARDNESS Vs DISSOLUTION PROTOCOL prepared and approved qYes qNo
OPERATIONAL PARAMETERS
1. FULL PRODUCTION FACILITIES qYes qNo
2. VALIDATION LOTS SIZE = COMMERCIAL SIZE qYes qNo
3. STANDARD PRODUCTION DOCUMENTATION PACKAGE qYes qNo
4. ROUTINE SOPS ; ROUTINE STAFF ; ROUTINE QC ; ROUTINE QA qYes qNo
SIMILARITY TESTING
1. Includes minimum of FIRST THREE commercial batch lots PER size qYes qNo
2. Minimum of FIRST THREE commercial batch lots PER dosage strength qYes qNo
3. Similarity Testing Performed on FIRST THREE commercial batches qYes qNo
4. Similarity Testing evaluated COMPARING commercial batch lots qYes qNo
(between themselves - intrabatch) and between the Pivotal Biostudy batch
lot as well (inter-batch comparison).
VALIDATION REPORTS
5. TEST RESULTS OF VALIDATION SAMPLING PLAN qYes qNo
6. TABULATION OF TEST RESULTS AND SUPPORTING GRAPHS qYes qNo
7. PRODUCT VALIDATION REPORT qYes qNo
ED. N0: 03 Effective Date: APPROVED:
Replaces 02
Ed. Status : DD / MM / Y2K
Current
Department R&D RA QC / QA
The validation requirements of an FDA approved generic drug product requires that the first three
consecutive commercial batches to be validated. The validation process must demonstrate that the
overall manufacturing process performs consistently, as expected, and that the generic drug product
consistently meets its predetermined filed specifications (i.e. in-process & finished product
specifications).
ØG u i d e l i n e s ×
CL # PVG-D03-Y2K
VALIDATION PROTOCOL
The Contents of a Validation Protocol
VALIDATION PROTOCOL - (Tablet & Capsule Forms)
n PURPOSE of DISSOLUTION STUDY ¨Yes ¨No
n RESPONSIBILITIES of LAB PERSONNEL ¨Yes ¨No
n FREQUENCY of Testing ¨Yes ¨No
n ANALYSIS - CRITICAL STEPS - DEFINED ¨Yes ¨No
n CRITICAL PARAMETERS - LIMITS & TARGET VALUES SPECIFIED ¨Yes ¨No
n SAMPLING PLAN - Written ¨Yes ¨No
n TESTING PLAN - Physical & Chemical ¨Yes ¨No
n ACCEPTANCE CRITERIA ¨Yes ¨No
ØG u i d e l i n e s ×
Sampling Record for Final Granulate
PRODUCT STRENGTH q- LAB RECEIPT No
Batch No: Date of Sampling: Sampling Unit No: Die No:
q PIVOTAL & q BIOEQUIVALENCE LOT q PROCESS QUALIFICATION LOT
q VALIDATION LOT q RE-VALIDATION LOT
Milled No of Weight of TOP MIDDLE BOTTOM Time Signature
Granulate Samples Sample - g
Final
Granulate
Choice of
FORMULA
PROCESS 1. LOD QUALIFICATION
OPTIMIZATION
2. LUBRICANT QUALIFICATION
FORMULATION
Drug Development Phase (VIA WRITTEN PROTOCOLS)
SCALE-UP
Choice of
PROCESS PROCESS QUALIFICATION
The Documentation
PROCESS OPTIMIZATION
PIVOTAL BATCH
VALIDATION - PROTOCOL
SAMPLING PLAN
TESTING PLAN
VALIDATION
3 Full Size Batches VALIDATION REPORT
MARKETED PRODUCT
RE-VALIDATION
of Commercial Product after;
MAJOR
CHANGE
[1] New Process Equipment Introduced
[2] Major Process Change.
INTRA-BATCH VARIABILITY
(Reproducibility of the
O P T I M I Z A T I O N
Three Validation Lots)
via:
• Dissolution Profile
• Content Uniformity
• Parameters within LCL-UCL Range SCALE-UP
MARKETED PRODUCT
CONTROL LIMITS
UCL
LCL
RE-VALIDATION
MAJOR of Commercial Product after;
PROCESS CAPABILITY CHANGE [1] New Process Equipment Introduced
CP ≤ 1 [2] Major Process Change.
TESTING PLAN
CORES COATED
Tablet Weight Tablet Weight
Thickness Thickness
Hardness & Hardness Qualification Profile -
Friability / Disintegration - (if necessary) -
Assay (composite 10 cores) Assay THREE
Content Uniformity Content Uniformity
Dissolution Dissolution
Dissolution Profile (Hardness Qualification) Dissolution Profile
VALIDATION REPORT
• Aim and Purpose of Study • Acceptance Criteria Evaluation
• List of Raw Material used in MNF • Analysis of Results (+ statistical) FOUR
• List of MNF Equipment (Pre-Qualified) • Validation Statement of Process
• Critical Process Steps Studied • Validation Recommendations
• Results of Data Collected • Attachments (Batch Records & CoAs)
INTRA-BATCH VARIABILITY
(Reproducibility of the
Three Validation Lots) VALIDATION BATCH
via:
• Dissolution Profile
• Content Uniformity
• Parameters within LCL-UCL Range
VALIDATION LOTS
Minimum 3 Full Size Batches
per strength and size
INTER-BATCH EQUIVALENCY
The Validation Documentation
Show Statistical Similarity
(Equivalency of the three Validation Lots
and the Biostudy Batch) VALIDATION - PROTOCOL
• Dissolution Profile
PROCESS CAPABILITY
MARKETED PRODUCT
CP ≤ 1
RE-VALIDATION
MAJOR of Commercial Product after;
CHANGE [1] New Process Equipment Introduced
[2] Major Process Change.
Pre-Approval
Inspections
'…Did you do what you wrote? - Can you do what you said?…'
Where the object is to routinely Firms who have open and strong
manufacture at an approved GMP US audit programs in place inevitably do
commercial production site then the well in PAIs. Multiple mock trail-runs
pivotal batches, for regulatory prior the anticipated inspection will
submission to the authorities, should iron-out the hidden deficiencies and
always be manufactured at the US give valuable training and confidence
commercial site as the intended market to all personnel concerned.
is the USA. What are Common
Conducting Mock Inspections. PAI
Preparing your department and firm’s Issues and Concerns ?
personnel for what is to come seems a
daunting challenge. It does not have to Staff learn to answer questions directly
be so. An intensive pre-approval in- - and not to talk any more than
house mock inspection with QA or RA necessary or advance gratuitous
role playing as the agency inspection information. Where foreign languages
team pays excellent dividends. are involved, a good practice is to
appoint a QA/RA spokesperson to
Conduct an Intensive handle all communication and remain
close to the inspectorate at all times.
mock In-house Create an SOP to define Agency-
Inspection Company Interactions and
communications. Key personnel speak
Challenge and dissect the through the designated QA and RA
manufacturing and controls necessary Director to the inspectorate. As an
to support approval of a new or inspection has generally two or
reformulated drug whether it be a new sometimes three agency personnel -
active or a generic. the need to appoint at least three
Asking awkward questions, before the Spokespersons e.g. QA, RA and
inspector does, may save months of Production usually cover all situations.
work and your companies reputation -
such as; Ensure the investigation
♦ Formula development runs smoothly
♦ Method development on-site
♦ Actual Process Development The firms representatives should each
♦ Master Specifications have a set of ‘runners’ - key aids, who
do not speak but rapidly retrieve
⇒ active documentation requested for by the
⇒ Raw Materials inspectors.
⇒ In-process
⇒ Finished Process Review rapidly the requested
⇒ Cleaning documentation with the Head of the
⇒ Check /Stability relevant department as to the
correctness of the data requested.
♦ Stability File Submission Batch
Then present the information to the
♦ Scale-up inspector by opening the desired
♦ Validation Lots page(s) - use book markers -
♦ Data Archiving (& Rapid Retrieval) highlighting the raw data only. Don’t
allow reviewers to wander through lab-
books.
PAI SUMMARY:
Set aside the board • Pharmaceutical Development
room or other venue Notebooks containing All the
experimental data
for the Inspectors ◊
exclusive use • Analytical Development Notebooks
containing all the analytical / micro
As lab books are confidential data from early active material
documents, inspectors do not and investigation to validation of stability
should not page randomly through an indicating assays and impurities
analytical raw data lab book. The ◊
opened lab book is placed in front of • A full Development Report based on
the reviewer with ‘begin and end’ a series of development lots in a
bookmarks designating the raw data for progressive, logical manner with
the relevant batch(es) requested. scientifically valid
Daily Work Venue decisions
Reserve the firms board room or a large being made
meeting room as a routine workplace
for the company and PAI Officials. ◊
Display all ANDAs, NDAs, Development • Full Plant Documentation of the
Reports, Protocols, cleaning and Pivotal Lot Manufacture
process validation reports. Copies of & Packing
the USP, BP and other Official ◊
Reference works used should be on • Full stability data and an operational
hand. & functional stability unit
Select what to file ◊
• The submitted ANDA is able to be
after the PAI - via SUPAC verified against the departmental
(post approval) raw data
◊
Display on a separate table all the • The plant is capable of
product documentation under review. producing the drug as specified in
Label and group documentation and the ANDA
reports together (e.g. ANDA +
Development Report + Cleaning and ◊
process validation protocols and • Full cGMP in place
Reports represent a product throughout the plant and labs.
documentation group). Lastly provide ◊
light refreshments. 3 • An operational and functional
SOP system
◊
• IQOQ Equipment (calibrated)
The Agency needs the
assurance ◊
• Process Validation Protocol
that product development prepared
yields the following:- è ◊
ØC H E C K L I S T ×
CL # HPGD-03-01Y2K
1. The firm has selected a multifaceted pre-approval inspection team? qYes qNo
2. The PAI Team has outlined its charter and team responsibilities ? qYes qNo
3. The Quality Assurance Head has approved and signed the charter ? qYes qNo
4. The PAI team has issued a SOP detailing regulatory procedures and qYes qNo
protocols to be followed during inspection procedures ?
5. The PAI team leader is the Head of Quality Assurance or his designate? qYes qNo
6. The PAI team is fully updated on the FDA Pre-approval Program ? qYes qNo
7. A suitably equipped room has been dedicated for use during the PAI? qYes qNo
8. A working list of documents, ANDA files, Pharmacopoeia, raw data lab qYes qNo
books etc. is available on-site in the PAI audit room, during the course
of the inspection ?
9. The PAI team has reviewed all product-specific files and appropriate qYes qNo
documentation for accuracy, clarity, and consistency - noting logical
dates, and signatures ?
10.The PAI team has focused on the pivotal batch dossier and the qYes qNo
proposed commercial manufacturing documentation with on-site
evaluations of actual filed manufacturing facilities?
11.The PAI team has, examined the product development report, together qYes qNo
with the project leader, and evaluated the explanations and scientific
rationale for changes that occurred during the process development right
up to the pivotal lot manufacture?
12.The PAI team has evaluated the product(s) cleaning validation, residual qYes qNo
limits, acceptance criteria and actual cleaning validation results?
13.The PAI team has evaluated the product(s) process validation protocol? qYes qNo
14.The PAI team is geared to provide assistance in the preparation of qYes qNo
written responses to the investigators concerning adverse finding -
during the actual PAI review?
15.The PAI team has established and coached a core of key personnel able qYes qNo
to properly respond to questions asked by FDA investigators during the
PAI review?
333
ØC H E C K L I S T ×
CL # HPGD-03-01Y2K
2. Development
3. Review the development report in detail and assure adequate qYes qNo
supporting documentation for process and optimization changes
made?
4. Has the active’s particle size, bulk density and polymorphism been qYes qNo
fully addressed?
5. Does the development report support a scientific basis for the qYes qNo
process validation protocol?
7. Process Qualification
9. Review cleaning validation, residual limits, and acceptance criteria qYes qNo
for the cleaning process?
10.Review that similar water quality was used from process qYes qNo
qualification to commercial lots?
11.Review that any reprocessing/rework stage has been fully qualified qYes qNo
and evaluated?
Ø CHECKLIST×
CL # HPGD-03-01Y2K
PIVOTAL BATCH
1. Review list of manufacturing and packaging equipment used in qYes qNo
pivotal lot.
2. Examine machine cards and cleaning verification cards or tags. qYes qNo
3. Examine room cleaning cards. qYes qNo
4. Examine all pivotal manufacturing, packaging and labeling records qYes qNo
(review date order, signatures present manufacturing deviations,
yield accountability vs. SOPs, sampling plan / retention samples) ?
5. Review the plan/protocol for sampling and testing the pivotal lot. qYes qNo
6. Examine intended production documentation. qYes qNo
7. Review Manufacturing Deviation Report relevant to each pivotal qYes qNo
batch.
8. Review conformance and compliance to manufacturing time qYes qNo
limitations.
9. Review excipient monographs vs. CoAs; routine and full testing qYes qNo
procedures and test dates?
10.Review overall disbursement of pivotal batch - e.g. QC release, qYes qNo
retention, stability, biostudy samples and full reconciliation of product
trail for each pack type used, and packs placed on stability?
11.Identify contract manufacturers or packages for documentation and qYes qNo
compliance auditing?
333
Ø CHECKLIST×
CL # HPGD-03-01Y2K
SCALE-UP TO COMMERCIAL
15. Review scale-up documentation and ensure all changes are within qYes qNo
SUPAC rules ?
16. Review ANDA formula and manufacturing variations are within qYes qNo
SUPAC limits?
17. Review side-by-side comparison of pivotal vs. commercial process, qYes qNo
facilities and equip.?
18. Review SOPs relating to issue and review of batch records, receipt, qYes qNo
handling and processing of raw materials, packaging components and
labeling, cleaning procedures, maintenance, HVAC, calibration and
equipment use, change control and manufacture.
19. Review site (manufacturing and labs) personnel training procedures qYes qNo
and training records
qYes qNo
PROCESS VALIDATION
20. Validation protocol for first three consecutive commercial lots on- qYes qNo
site ?
21. Review the side-by-side comparison between pivotal and validation qYes qNo
equipment /facilities ?
22. Review the protocol for sampling and testing the validation lots qYes qNo
(part of validation protocol)
ANALYTICAL
23. Perform a thorough check on the lab notebook referencing all qYes qNo
pivotal tests ?
24. Review method validation for in-house assays, impurities and qYes qNo
dissolution test?
25. Review stability-indicating assay and the impact of any placebo qYes qNo
effect ?
26. Review all out-of-specification results (log) on analytical or product qYes qNo
failures?
ØC H E C K L I S T ×
CL # HPGD-03-01Y2K
STABILITY
27. Does pivotal batch remains in check specifications after 3 months qYes qNo
accelerated (40oC) testing ?
28. Have samples been placed on stability within 30 days of qYes qNo
manufacturing release CoA ?
29. Insure stability testing is performed according to stability protocol ? qYes qNo
30. Statement on proposed expiration period in accordance with qYes qNo
obtained test results ?
31. Review all stability data, in-out dates, exposure times, temperatures qYes qNo
and humidity (RH) including control of environmental parameters
(recording devices, graphs and any OOS conditions) ?
32. Review Stability SOPs and equipment; Are chambers, probes and qYes qNo
monitors calibrated ?
MICROBIOLOGICAL
33. Microbial parameters in compendial excipients are retested every qYes qNo
12 months ?
34. Preservative containing excipients are tested for preservative qYes qNo
efficacy ?
35. Appropriate use of validated in-house methods with proper method qYes qNo
controls ?
36. Review of microbial testing facilities, test methods, calibration and qYes qNo
laboratory SOPs?
37. Full GMP review (on-site/mail audit) of contract facilities used ? qYes qNo
38. Review of analytical testing facilities methods, validation and qYes qNo
laboratory SOPs ?
39. All facilities listed in file submission are fully capable of performing qYes qNo
designated tasks ?
40. Review of packaging and labeling procedures, in-process controls qYes qNo
and SOPs ?
333
The principle established in ICH Q1A -- kinetic temperature in any region of the
that information on stability generated in world can be derived from climatic data.
any one of the three areas of the EU, (Grimm, W., Drugs Made in Germany, 28:196-202,
1985, and 29:39-47,1986). Referenced in [ICH Q1A]
Japan, and the USA would be mutually
acceptable in both of the other two areas The recommendations in this guidance
--- is incorporated in this guidance are effective upon publication of the
document. final guidance and should be followed in
In fact, much of the text of the guidance preparing new applications, re-
on drug substances and drug products is submissions, and supplements.
incorporated directly from the ICH Q1A This guidance represents FDA’s current
text (e.g. II.A. and II.B.). thinking on how the stability section of
The guidance is intended to replace the drug and biologics applications should
be prepared. An applicant may choose to
12 year old, Guideline For Submitting
use alternative procedures.
Documentation for the Stability of Human
Drugs and Biologics, published in If an applicant chooses to depart from
February 1987. the recommendations set forth in this
guidance, the applicant is encouraged to
It applies to all drug substances and discuss the matter with FDA prior to
products submitted to the Center for initiating studies that may later be
Drug Evaluation and Research (CDER). determined to be unacceptable.
This guidance also applies to biological
FDA recognizes that the time necessary
products that are included in the scope
for applicants to establish new
of the ICH Q5C Stability Annex, Stability
procedures, install, and commission the
Testing of Biotechnology Drug Products
new temperature and relative humidity-
(July 1996) and all other products
controlled rooms / cabinets, carry out
submitted to the Center for Biologics
appropriate stability studies on batches
Evaluation and Research (CBER).
of product, and submit the information in
The draft guidance provides an application may prevent some
recommendations for the design of applicants from generating data
stability studies for drug substances and consistent with the recommendations in
drug products that should result in a the guidance for some time.
statistically acceptable level of However, since this guidance represents
confidence for the established retest or FDA’s current thinking, recommendations
expiration dating period for each type of regarding stability data submission not
application. conforming with this guidance is possible
The applicant is responsible for with justification.
confirming the originally established Applications withdrawn prior to
retest and expiration dating periods by publication of this guidance should not
continual assessment of stability normally have to include stability data in
properties (21 CFR 211.166). conformance with the guidance upon
resubmission. However, if new stability
Continuing confirmation of these dating studies are conducted to support the
periods should be an important submission, such studies should be
consideration in the applicant’s stability conducted as per guidance.
program.
The choice of test conditions defined in A comprehensive glossary is included,
this guidance is based on an analysis of and contains definitions of the major
the effects of climatic conditions in the terms and origins of the definitions
EU, Japan, and the USA. The mean derived either from ICH, CFR, or USP.
The batch(es) used for stability testing appropriate expiration dating period is
should comply fully with the proposed determined.
specifications for the product and be Extension of the tentative expiration
packaged in the market package, and
dating period should be based on data
the release assay should be within
generated on at least three production
reasonable variation (taking into account
batches tested according to the
inherent assay variability) from the
approved protocol outlined in the stability
labeled strength or theoretical strength of
commitment. Reporting of the data
the reference listed drug.
should follow Section VI. of this guidance
Overages must mimic ANDA STABILITY
the RLD Product COMMITMENT on THREE
If formulated with an overage, the commercial lots until long
overage should be justified as necessary term testing is complete
to match that of the reference listed ANDAs withdrawn prior to publication of
drug.
this guidance should not normally have
Other supportive stability data may be
to include stability data in conformance
submitted on drug product batches that
with the guidance upon resubmission if
may or may not meet the above criteria.
the original application was withdrawn
In Modified Release due to non-stability related issues.
However, if new stability studies are
Solid Dosage Forms conducted to support the submission,
the PQ batch can be such studies should be conducted as
recommended in the guidance.
the third stability batch
Data on relevant research batches, NEW RULES SUMMARY
investigational formulations, alternate
container/closure systems, or from other
related studies may also be submitted to SIMPLE ANDAs
support the stability of the drug product. Ø One batch of 100 000 (net) for 1, 2, &
The supportive stability data should be 3 months at 40oC ± 2oC / 75% RH ± 5%.
clearly identified. Ø Long Term Study started i.e. 3 or 6
months - whatever is available.
[G] Stability Study Acceptance
Ø Commitment to THREE production
If the results are satisfactory, a tentative
lots until long term data complete.
expiration dating period of up to 24
months at the labeled storage conditions Ø Significant Change clause effective.
may be granted. COMPLEX ANDAs
Where data from accelerated studies Ø TWO batches of 100 000 net for 1, 2,
are used to project a tentative expiration & 3 months at 40oC ± 2oC / 75% RH ±
dating period that is beyond a date 5% PLUS Small scale (e.g. PQ) lot.
supported by actual long-term studies on Ø Long Term Study started i.e. 3 or 6
production batches, then the application months - whatever is available.
should include a commitment to conduct Ø Commitment to THREE production
long-term stability studies on the first lots until long term data complete.
three production batches and annual Ø Significant Change clause effective.
batches until the tentative expiration
dating period is verified, or the
TABLE OF CONTENTS
The following example highlights the Overall ANDA Guideline Requirements for the
Stability Section of an application. Only two out of the required sixteen stability
tabulations (stability profiles) are shown. A separate profile is required for each
strength, pack size and challenge temperature and humidity. The stability section is
divided into seven essential parts, as follows:
17.3 Stability Protocol for Post Approval Production Batches (This is the ANDA
stability commitment on what will be done after approval of the file)
17.4 Individual Stability Reports (stability profiles) indicating results obtained from
the Pivotal lot after [3] months accelerated and [X] months controlled room
temperature / humidity studies
17.5 Package Configuration and sizes (largest and smallest) used in stability
studies.
17.6 Stability Protocol used for (two) Pivotal Batch lots consisting of one
PIVOTAL Lot for each marketed strength.
17.7 Stability Data Summary Report (plus graphical presentations, graphs of assay
values vs. time ).
Overview
Stability testing is performed on each strength of three batches in the largest and
smallest container-closure systems proposed for marketing; i.e. in each material type,
namely plastic (HDPE/HDPP), or glass packs.
When more than one closure for the same container material type (e.g. HDPE
container) is used in the proposed marketing containers, the largest and smallest
container-closure configuration is tested, - for both accelerated and long term studies.
In cases where plastic bottles of the same size range and shape are manufactured
from different thermoplastic resins, they exhibition different storage characteristics and
thus are considered as completely separate container-closure systems.
The number of stability tests conducted can be quite large in such cases. The example
below for the following packaging configuration highlights the number of stability tests
needed. Tests can be significantly reduced using a matrix stability protocol.
1. HDPE (s/s & l/s) container with inner liner and HDPE CRC cap.
2. HDPE (s/s & l/s) container with inner liner and metal cap.
A ll stability data support the proposed expiration period of 2 years when the product
is stored at room temperatures.
Stability commitment
Long term commercial stability studied in accordance with the approved stability
protocol shall be carried out by [Generic Company Name Inc. / Ltd.] The stability
results of these studies shall be submitted in the annual ANDA Reports filed on the
anniversary date of the submitted product.
[Generic Company Name Inc. / Ltd.] commits to remove any batch promptly from the
market place any material falling outside the products check specifications.
Extensions to the expiration date will be made via the annual ANDA Reports as
acceptable long term stability data is obtained,
Rework procedures may be submitted for batches that fail to meet established specifications. Prior
to implementation, these procedures will be submitted in a supplement in accord with: 21 CFR
314.70 (b)(2)(x) on a lot by lot basis.
Report Format
Results will be tabulated in the format of the Stability Report Form:
1) Product Name, and Strength 9) Stability Start Date
2) Batch Number and Batch size 10) Manufacturing Site
3) Storage Conditions and Intervals 11) Manufacturer of Bulk Drug
4) Container/Closure Systems - Description 12) Inventory Control Number of (11)
5) Inventory Control Number of (4) 13) Manufacturer of Container/Closure
6) Fill Size and No of units on stability 14) Formulation
7) Batch Manufacturing Date 15) Data profile
8) Batch Packaging Date 16) Methodology and Specifications
The data indicate that the formulation is stable, with no observed degradation peaks,
under test conditions. There were no significant changes in either the physical
chemical, or microbiological specifications in any samples evaluated after the exposed
storage test conditions.
The attached tables and graphs are summaries of the results for the parameters used
to establish the stability profile of [Generic name] TABLETS [USP] [000.0] mg. for the
pivotal batch per dosage strength, where applicable.
Included, are assay chromatogram spectra of the stability tests for zero time (T0) and
the three (3) months accelerated test conditions.
[Generic name] TABLETS [USP] [000.0] mg were stored at accelerated conditions
(40o ±2 o C / 75% RH ± 5%) and at room temperature (25° ±2°C/ 60% RH ±5%) in the
proposed market container/closure system. All stability data supports the proposed
expiration period of 2 years when the product is stored at room temperatures.
SIGNIFICANT CHANGE CLAUSE:
Where PRODUCTS fail the accelerated testing, Intermediate testing is performed
according to the following specification.
Intermediate testing 300 ±20C/60 ±5%RH
0,1,2,3,6, 9 and 12 months
2
Glass Container Closure Liner System
ý ý
ý ý
Bulk Packaging
ý ý
Number of Pivotal Batches ('100 000+' rule) [1]
New for
Number of NON-Pivotal Batches (i.e. PQ Lot) [0]
MR
TOTAL NUMBER OF STABILITY BATCHES 1
Dosage
Number of Temperature/RH Levels 2 Forms
Number of Dosage Strengths 2 only
Number of Container-Closure Sizes 2
Number of Resins present in Containers 1
Number of Closure Systems 2
Number of Resins present in Closures 1
Number of Stability Studies Performed 16
Number of resins used in the HDPE containers = one resin from same supplier.
1
Typical Assembly : HDPE container with HDPE CRC / METAL SCREW CAP and inner LINER.
2
Typical Assembly : Glass bottle with HDPE CRC / METAL SCREW CAP and inner LINER
STABILITY REPORT
1 Product name, dosage form and strength. Generic name] [000] mg.
2 Fill size 000 [USP]
3 Site of Manufacture NJ MNF SITE
4 Batch or lot number 000
5 Batch size (type) 000 000
6 Batch manufacturing Date and Packaging Date Month DD, 199Y / Month DD, 199Y
7 Manufacturer of Active Material (approved supplier) LEK Chemical Co. dd
8 Date placed on stability Month DD, 199Y
9 Batch number or receiving number of Active Material LK 2323
10 Full details of container/closure system (type, material, resin) 100cc HDPP (LR-7340-43) CRC
HDPP white cap (resin LR-7340-43)
11 Goods Receiving number of container-liner GRN 96-2-02234 (body)
Goods Receiving number of closure GRN 96-2-02237 (cap)
12 Manufacturer of container/closure Wheeler Cap Co PA USA.
13 Objective of the stability program þ Pivotal Batch
14 Site where stability test conducted US PA MAN Site
15 Number of units to be sent for testing in each time interval 2 x 000
16 Analytical method number and Edition Number for each stability indicating test S-I 555-03 / S-I 1234 Ed . 03
17 Stability specifications indicating names of test required. Tabulated
18 Number of packages placed on stability 70
19 Testing intervals required 0, 1, 2, 3 (6) months
20 Stability storage conditions 40 degrees C / 75% RH
HPLC/TLC
Period Date of Contents ASSAY DISSOLUTION
Analysis Appearance Impurity
% Profile Percentage of label claim dissolved in 45 minutes
Impurities I
White to
Month 90.0 % - Impurities II NLT 80% (Q) in 45 minutes
Creamish
110.0% Impurities III
Powder
Total < 3.0
Method # S-I-000 -01 S-I-000 -02 S-I 000 -03 S-I 000-04 Mean C.V.
0 1/6/97 conforms 100.3 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
3 5/9/97 conforms 99.8 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
6 9/12/97 conforms 101.3 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
9 15/3/98 conforms 101.4 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
STABILITY PARAMETERS
21. Product Formula On Stability (Formula No: 000- Identical to ANDA Sections 6, 7, 11, & 12
1. Active material BP 7. Anhydrous Lactose NF 230.00
2. Povidone USP 9.00 8. Magnesium Stearate NF 3.00
3. Colloidal Silicon Dioxide NF 2.10 9. Opadry OY-S-20000 (light Color)
4. Starch NF 5.30
5. Starch NF (Redried) 27.00
6. Purified Water USP NF (Processing Solvent Only)
STABILITY REPORT
1 Product name, dosage form and strength. [Generic name] [000.0] mg.
2 Fill size 00 [USP]
3 Site of Manufacture NJ MNF SITE
4 Batch or lot number P-5432
5 Batch size (type) 000
6 Batch manufacturing Date and Packaging Date Month DD, 1998 / Month DD, 1998
7 Manufacturer of Active Material (approved supplier) LEK Chemical Co. dd
8 Date placed on stability June 15, 199Y
9 Batch number or receiving number of Active Material LK 2323
10 Full details of container/closure system (type, material, resin) 200cc HDPP (LR-7340-43) CRC
HDPP white cap (resin LR-7340-43)
11 Goods Receiving number of container-liner GRN 96-2-02234 (body)
Goods Receiving number of closure GRN 96-2-02237 (cap)
12 Manufacturer of container/closure Wheeler Cap Co PA USA.
13 Objective of the stability program þ Pivotal Batch
14 Site where stability test conducted US PA MNF Site
15 Number of units to be sent for testing in each time interval 2 x 00
16 Analytical method number and Edition Number for each stability indicating test S-I 555-03 / S-I 1234 Ed . 03
17 Stability specifications indicating names of test required. Tabulated
18 Number of packages placed on stability 70
19 Testing intervals required 0, 3, 6, 9, 12, 18, 24, 36… months
20 Stability storage conditions 25 degrees C / 60% RH
Contents HPLC/TLC
Period Date of Appearance DISSOLUTION
Analysis ASSAY Impurity
ID
% Profile Percentage of label claim dissolved in 45 minutes
Impurities I
White to
Month Lab book 90.0 % - Impurities II NLT 80% (Q) in 45 minutes
Creamish
Ref. No 110.0% Impurities III
Powder
Total < 3.0
Method # S-I-000 -01 S-I-000 -02 S-I 000 -03 S-I 000-04 Mean C.V.
0 1/6/97 conforms 100.3 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
L/Book 30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
N0: ___
Page___ 45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
3 5/9/97 conforms 99.8 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
L/Book 30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
N0: Page
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
6 9/12/97 conforms 101.3 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
L/Book 30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
N0: Page
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
9 15/3/98 conforms 101.4 conforms 15: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
L/Book
N0: Page 30: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
45: 00.0; 00.0; 00.0; 00.0; 00.0; 00.0 00.0 0.0
STABILITY PARAMETERS
Additional Tests performed Annually: Microbial Limit Test USP and Residual Solvents.
21. Product Formula On Stability (Formula No: 000- Identical to ANDA Sections 6, 7, 11, & 12.
1. Active material BP 7. Anhydrous Lactose NF 230.00
2. Povidone USP 9.00 8. Magnesium Stearate NF 3.00
3. Colloidal Silicon Dioxide NF 2.10 9. Opadry OY-S-20000 (light Color)
4. Starch NF 5.30
5. Starch NF (Redried) 27.00
6. Purified Water USP NF (Processing Solvent Only)
Container Drug Plastics & Drug Plastics & Drug Plastics & Drug Plastics &
manufacturer Glass Co. Inc. Glass Co. Inc. Glass Co. Inc. Glass Co. Inc.
Container size 00 / 000cc round, 00 / 000cc round, 00 / 000cc round, 00 / 000cc round,
White HDPE White HDPE White HDPE White HDPE
container container container container
Resin Type HDPE HDPE HDPE HDPE
Quantum LR-7340-43 Quantum LR-7340-43 Quantum LR-7340-43 Quantum LR-7340-43
Cap U.S. CAN U.S. CAN U.S. CAN U.S. CAN
Manufacturer (Penn-Wheeling (Penn-Wheeling (Penn-Wheeling (Penn-Wheeling
Closure Corp.) Closure Corp.) Closure Corp.) Closure Corp.)
11087 PE White Ampacet White Ampacet White Ampacet White Ampacet
White Master 11078 Polyethylene 11078 Polyethylene 11078 Polyethylene 11078 Polyethylene
batch
Cap Type ROPP HDPE Cap ROPP HDPE Cap ROPP HDPE Cap ROPP HDPE Cap
CRC HDPE Cap CRC HDPE Cap CRC HDPE Cap CRC HDPE Cap
Cap Size 00 / 00 mm 00 / 00 mm 00 / 00 mm 00 / 00 mm
Closure Liner Tekni-Plex Inc. Tekni-Plex Inc. Tekni-Plex Inc. Tekni-Plex Inc.
Foam seal Mfg Foamseal PS 22 Foamseal PS 22 Foamseal PS 22 Foamseal PS 22
Inner liner TEKNISEAL RVT TEKNISEAL X-14 TEKNISEAL RVT TEKNISEAL X-14
composition + LF (polyethylene/Kraft + LF (polyethylene/Kraft
Paper laminate) Paper laminate)
CONTAINER Lot #00000 Lot #00000 Lot #00000 Lot #00000
CoA #0000 CoA #0000 CoA #0000 CoA #0000
CONTAINER Lot #00000 Lot #00000 Lot #00000 Lot #00000
CoA #0000 CoA #0000 CoA #0000 CoA #0000
CAP Lot #00000 Lot #00000 Lot #00000 Lot #00000
CoA #0000 CoA #0000 CoA #0000 CoA #0000
CAP Lot #00000 Lot #00000 Lot #00000 Lot #00000
CoA #0000 CoA #0000 CoA #0000 CoA #0000
LINER Lot #00000 Lot #00000 Lot #00000 Lot #00000
CoA #0000 CoA #0000 CoA #0000 CoA #0000
SEAL Lot #00000 Lot #00000 CoA #00000 CoA #00000
CoA = Certificate of Analysis/Compliance.
seen after 3 months during the determined based on the available data
accelerated stability study. from the additional studies)
Ü The tentative expiration dating Complex Dosage Forms
period will be determined based on the
available data from the additional study. MR/CR/ER/DR/MDIs
×COMPLEX DOSAGE FORMSØ
Ø require THREE
Exceptions to the ANDA Data Package
Recommendations.
Stability batches
The following may be considered Data Package for Approval
exceptions to the general ANDA
recommendations:- ◊ Full-term stability testing of the
primary stability batch(es) is suggested.
Complex dosage forms, such as However, in the absence of full-term
Ü Modified-release products. stability data for the drug product,
Ü Transdermal patches. adequate accelerated stability data
Ü Metered-dose inhalers. combined with available long-term data
can be used as the basis for granting a
Ü Drug products without a tentative expiration dating period.
significant body of information.
Ü New dosage forms submitted
◊ The batch(es) used for stability
testing should comply fully with the
through the ANDA suitability petition
proposed specifications for the product
process
(Other exceptions may exist and should be
and be packaged in the market
discussed with the Office of Generic Drugs. package, and the release assay should
Refer Q1C applications). be within reasonable variation (taking
An ANDA that is determined to be one into account inherent assay variability)
of the above categories should contain from the labeled strength or theoretical
a modified ICH Q1A stability data strength of the reference listed drug.
package, including: ◊ If formulated with an overage, the
◊ 3-month accelerated stability studies. overage should be justified as
necessary to match that of the
◊ Long-term stability studies (available reference listed drug.
data at the time of original filing and
subsequent amendments). The ◊ Other supportive stability data may
expiration dating period for complex be submitted on drug product batches
dosage forms will be determined based that may or may not
on available long-term stability data ◊ meet the above criteria. Data on
submitted in the application. relevant research batches,
◊A minimum of three batches investigational formulations, alternate
manufactured in accordance with the container / closure systems, or from
ICH Q1A batch size recommendations other related studies may also be
(refer Section II B of this guidance -NDA Drug submitted to support the stability of the
Products or to V.B. of the ICH Q1A guidance). drug product. The supportive stability
Additional stability studies (12 months data should be clearly identified.
at the intermediate conditions or long-
term stability testing through the
Stability Study Acceptance
proposed expiration date) if significant If the results are satisfactory, a
change is seen after 3 months during tentative expiration dating period of up
the accelerated stability studies (the to 24 months at the labeled storage
tentative expiration dating period will be conditions may be granted.
Where data from accelerated studies Additional data from ongoing studies
are used to project a tentative and regular annual batches should be
expiration dating period that is beyond included in the application’s annual
a date supported by actual long-term report. Annual reports should include
studies on production batches, the new or updated stability data generated
application should include a in accordance with the approved
commitment to conduct long-term stability protocol.
stability studies on the first three The data may include accelerated and
production batches and annual batches long-term studies for each product to
until the tentative expiration dating satisfy the standard stability
period is verified, or the appropriate commitment made in the original or
expiration dating period is determined. supplemental application, including the
Final Expiration Dating annual batch(es), and to support post-
approval changes.
means long term The data should be presented in an
stability studies on organized, comprehensive, and
three production lots cumulative format.
CONTENT OF STABILITY REPORTS
For Complex Dosages It is suggested that stability reports
Extension of the tentative expiration include the following information and
dating period should be based on data data to facilitate decisions concerning
generated on at least three production drug product stability:
batches tested according to the
GENERAL PRODUCT INFORMATION
approved protocol outlined in the
stability commitment. • Product Name
Reporting of the data should follow • Source
Section VI format of the guidance which • Manufacturing site(s)
applies to all application types • Date of manufacture of drug
ANDAs withdrawn prior to publication substance and drug/biological product.
of this guidance should not normally • Dosage form and strength,
have to include stability data in
conformance with the guidance upon • Product formulation.
resubmission if the original application • Composition, type, source, size, and
was withdrawn due to non-stability adequate description of container and
related issues. closure., seals, and desiccants (cotton
However, if new stability studies are coil / stuffers) should be identified.
conducted to support the submission, The application should provide a table
such studies should be conducted as of specific formulations under study.
recommended in the guidance. When more than one formulation has
been studied, the formulation number is
REPORTING STABILITY DATA acceptable.
A. General
Stability data should be included in the SPECIFICATIONS AND TEST
ANDA application (or supplement) they METHODOLOGY INFORMATION
are intended to support. The extent of • Physical, chemical, and
stability data expected at the time of microbiological regulatory specifications
submission is discussed at length and attributes/limits (or specific
throughout the guidance. references to ANDA or USP).
ANDA à STABILITY
DRUG PRODUCT à FLOWCHART
SIMPLE DOSAGE FORMS COMPLEX DOSAGE FORMS
× IR Ø × CR/MR/ER/DR/MDIs…Ø
Formulation for Simple Dosage Forms Formulation for Complex Dosage Forms
Immediate Release Solids & Liquids Modified Release Solids & MDIs
When Applicable
1 2 3 4 +5
Dissolution
Initial To Specification Drug Product
Limit of any exceeds 12
Assay decreases exceeds it pH
drug product dosage unit
> 5% during limits during
degradant is o specification
1, 2, or 3 months the 30 or 40 C or
exceeded during Study
of 30/40o C Study o Appearance
30/40 C Study
Initial assay must be or
'close to 100% rule' Physical
SIGNIFICANT CHANGE properties
IMPACT ON TESTING
Ü Longer times required for overall stability study evaluation - more costly studies.
Ü Formulations to be rugged / robust to withstand potential 5% assay change.
Ü Enhanced antioxidant / chelating formula optimization studies1 necessary
during product development stages.
Ü Tablet Hardness Qualification Study 2
essential in either PQ or Pivotal Batch
lots.
Ü Full Analytical Assay Validation must be performed at the PQ stage or earlier to
sufficiently evaluate potential significant change parameters.
Courtesy: International Journal of Generic Drugs, Volume 01, No 08 ,1997.
Courtesy: International Journal of Generic Drugs, Volume 01 ,No 06, 1997.
PURPOSE
[3]. The supervisor and the analyst shall then conduct a laboratory investigation
which shall include a review of the analytical principles using the “Checklist For
Laboratory Investigation Report” (See “Out of Specification Result SOP.”)
[4]. Where the laboratory investigation reveals that the cause of the questionable
result is a LABORATORY ERROR, i.e. one of the principles mentioned in the
checklist was at fault, the supervisor shall invalidate the original test results. The
supervisor shall record the conclusions and scientific rationale in the analyst's
laboratory notebook. The comments shall be signed and dated.
[5]. Where the cause is found to be faulty analytical technique, the test is repeated
(in duplicate from the beginning) on the same sample (i.e. from the same stability
sample container initially used for sampling and testing).
Ü If the sample passes the retest, then the result may be released and accepted.
Ü In cases of retest failure, proceed as per paragraph [9].
[6]. Where the cause is due to faulty analytical methodology, a new edition
methodology revision shall be prepared.
[7]. Where the laboratory investigation is INCONCLUSIVE, and the cause cannot
definitely be ascribed to laboratory error, proceed as follows:
[8]. If the relevant pharmacopoeia specifies acceptance criteria guidelines for the
particular type of test involved (dissolution, microbial limits etc.), the analyst shall
proceed with the testing according to the official method. If the retest passes, it is
reported according to the pharmacopoeial requirements. If the compendial retest
fails, the Quality Assurance Unit inform for further investigation, where appropriate.
[9]. Where, the relevant pharmacopoeia does not specify retesting procedures for
the particular type of test involved, TWO re-tests will be performed by TWO analysts
(that is, the sample shall be tested in duplicate).
The final result is calculate as the average of the THREE analysis (e.g. 6 results
which include the results from two re-tests and the original test result). No individual
analysis of the retest results shall fail the specifications.
[10]. In cases that the results are within the specified limits, the results are accepted.
[11]. If the retest result still remains questionable, the supervisor will inform the
Quality Assurance Unit for further investigation, if so required.
[12]. The Quality Assurance Unit shall fully review the data and decide if the test
results should be reported as is, or additional action is required.
CORRECTIVE ACTION
Retesting is performed on the same sample container originally used, (obtaining
new samples, new sample stock or performing new sampling procedures from bulk
material or stability stock is prohibited.)
DOCUMENTATION
Laboratory investigations shall be fully documented, and the conclusions signed and
dated by the Laboratory Manager.
In case where the investigation is passed on to the Quality Assurance Unit Manager,
an Investigation Report shall be prepared.
Immediate Notification
Investigation Started
LABORATORY INVESTIGATION
FINDINGS in LOGBOOK
PASS
FAIL
PASS
FAIL
STABILITY TESTING
P hotostability
in New Drug Products
‘…evaluating photostability is foremost for new chemical entities only - not in
g e n e r i c d r u g s , p r o v i d e d t h e c o n t a i n e r - c l o s u r e p r o t e c t i o n i s t h e s a m e …'
emission between 350 nm and 370 nm; studies, the samples should be in
a significant proportion of UV should be chemically inert and transparent
in both bands of 320 to 360 nm and 360 containers.
to 400 nm. [ICH Q1B] In these forced degradation studies, a
variety of exposure conditions may be
PROCEDURE [ICH Q1B] used, depending on the photosensitivity
For confirmatory studies, samples of the drug substance involved and the
should be exposed to light providing an intensity of the light sources used.
overall illumination of not less than 1.2 For development and validation
million lux hours and an integrated near purposes, it is appropriate to limit
ultraviolet energy of not less than 200 exposure and end the studies if
watt hours/square meter to allow direct extensive decomposition occurs.
comparisons to be made between the
drug substance and drug product. For photostable materials, studies may
be terminated after an appropriate
Samples may be exposed side-by-side exposure level has been used.
with a validated chemical actinometric The design of these experiments is left
system to ensure the specified light to the applicant’s discretion although the
exposure is obtained, or for the exposure levels used should be justified.
appropriate duration of time when Under forcing conditions, decomposition
conditions have been monitored using products may be observed that are
calibrated radiometers/lux meters. unlikely to be formed under the
An example of an actinometric conditions used for confirmatory studies.
procedure is provided. This information may be useful in
If protected samples (e.g., wrapped in developing and validating suitable
aluminum foil) are used as dark controls analytical methods.
to evaluate the contribution of thermally
induced change to the total observed If in practice it has been demonstrated
change, these should be placed they are not formed in the confirmatory
alongside the authentic sample. [ICH studies, these degradation products
Q1B]. need not be examined further.
Confirmatory studies should then be
undertaken to provide the information
DRUG SUBSTANCE [ICH Q1B] necessary for handling, packaging, and
For drug substances, photostability labeling (see Presentation of Samples,
testing should consist of two parts: for information on the design of these
Forced degradation testing and studies Section VIII.J.3., Procedure, and
confirmatory testing. 4.a. in guidelines).
The purpose of forced degradation Normally, only one batch of drug
testing studies is to evaluate the overall
substance is tested during the
photosensitivity of the material for
development phase, and then the
method development purposes and/or
photostability characteristics should be
degradation pathway elucidation.
confirmed on a single batch selected as
This testing may involve the drug described in the parent guidance if the
substance alone and/or in simple drug is clearly photostable or
solutions / suspensions to validate the photolabile.
analytical procedures. In these
Some adjustment of testing conditions specifications during the shelf life (Ref.
are when testing large volume ICH stability and impurity guidance).
containers (e.g., dispensing packs). QUININE CHEMICAL ACTINOMETRY
[b.] Analysis of Samples Details of an actinometric procedure for
monitoring exposure to a near UV
At the end of the exposure period, the
fluorescent lamp, based on the draft
samples should be examined for any
guidance are provided in the attached
changes in physical properties (e.g.,
Standard Operating Procedure.
n appearance
ACCEPTABLE & UNACCEPTABLE
n clarity of solution
PHOTOSTABILITY CHANGE
n color of solution
n dissolution / disintegration for dosage The extent of the drug product
forms such as capsules) photostability testing depends on the
n assay change that has occurred a the end of
n degradants each test tier described in the Decision
Suitably validated methods for Flow Chart for Photostability.
degradants likely to arise from Testing of Drug Products.
photochemical degradation processes Test results that are outside the
should be used. proposed acceptance criteria for the
product would not be considered
When powder samples are involved, acceptable change.
sampling should ensure that a
representative portion is used in This is a stress test designed to
individual tests. determine the intrinsic photostability
characteristics of new drug substances
For solid oral dosage form products, and products, and no correlation has
testing should be conducted on an been developed to equate a within
appropriately sized composite of, for specification result to an expiration
example, 20 tablets or capsules. Similar dating period.
sampling considerations, such as
homogenization or solubilization of the Evaluate Light Stress and Heat
entire sample, apply to other materials Stress Degradation Separately
that may not be homogeneous after
exposure (e.g., creams, ointments,
The acceptability of any observed
changes should be justified in the
suspensions).
application. It may be important to
The analysis of the exposed sample is consider other degradative processes
performed concomitantly with that of any (e.g., thermal) when justifying a
protected samples used as dark controls photostability change as acceptable
if these are used in the test. because the processes may be
[c.] Judgement of Results independent and additive.
Depending on the extent of change, For example, a 5 percent loss in
special labeling or packaging may be potency due to photodegradation may
needed to mitigate exposure to light. be considered acceptable if that is the
When evaluating the results of only type of degradation observed.
photostability studies to determine If the product is also expected to
whether change due to exposure to light degrade 5 percent over the shelf-life
is acceptable, it is important to consider due to thermal degradation, the photo-
the results obtained from other formal degradation may then be considered
stability studies to ensure that the unacceptable based on the potential
product will be within proposed additive effect of the changes.
In this case, precautions should be the product being removed from the
taken to mitigate the product’s exposure immediate package during the
to light. distribution process.
n For those products that are unlikely to
Under the intense light exposure be removed from the immediate
conditions included in the Q1B guidance, container, such as creams or ointments
certain colors in solid dosage forms may in tubes dispensed directly to the
fade. patient, and ophthalmic products, the
Quantitative analysis of the color change use of a labeling storage statement
is not recommended as these changes regarding light is optional.
are not likely to occur under actual
storage conditions. n For products that may be removed
In the absence of change in other from the immediate pack, such as
parameters such as assay, these color pharmacy bulk packs, a light storage
changes may be acceptable. statement should be included such as
“PROTECT FROM LIGHT. Dispense in
Two Standardized labeling a light-resistant container.”
instructions recommended to Change after exposure in the market
PROTECT FORM LIGHT pack:
PHOTOSTABILITY LABELING If changes that are observed are
acceptable only when the product in the
The data generated using the procedure market pack is exposed under the
described in the ICH Q1A guidance is conditions described in the Q1B
useful in determining when special guidance, labeling storage statements
handling or storage statements regarding light should be included.
regarding exposure to light should be
included in the product labeling (21 CFR
Examples of typical labeling cautions for
single-dose and multiple-dose products
201.57(k)(4)).
The labeling guidance provided below pertains respectively are either,
only to products as packaged for distribution.
Labeling Caution - Type Single Dose
Instructions and stability statements that may be
needed to address in-use conditions pursuant to
21 CFR 201.57(j) are not covered. n “PROTECT FROM LIGHT.
Retain in carton until time of use.”
Change after direct exposure:
If changes that are observed when the Labeling Caution - Type Single Dose
product is directly exposed under the
light conditions described in the Q1B n “PROTECT FROM LIGHT.
guidance are acceptable, no labeling Retain in carton until contents are used.”
storage statement regarding light is
needed. SOPS and 'Guidance for Industry'
n Violating an in-house SOP is a GMP
Change after exposure in the
violation and you can be cited for SOP
immediate container/closure: violations.
If changes observed when the product is n Violating a agency 'Guidance for
directly exposed are unacceptable, but Industry' in NOT a GMP violation and you
are acceptable when the product is cannot be cited for a violation.
tested in the immediate container / n Violating an in-house SOP which is
closure under the conditions described based on a 'Guidance for Industry' is a
in the Q1B guidance, the inclusion of a standard GMP violation.
labeling storage statement regarding The photostability SOP supplied is based
light would depend on the likelihood of on the current draft guidelines.
Formula
Change may
include OPAQUE
film coatings or
capsule shells Start of
Decision Tree
Drug Product
spread in a
CHANGE
mono layer
FORMULA YES glass petridish
Dosage Form
(Coating) Directly Exposed
NO
Drug Product in
Primary
CHANGE container-
Dosage Form in
Primary closure system
Primary Pack (without carton)
Pack YES
NO
CHANGE Drug Product
Marketing Dosage Form in in Primary &
Pack Marketing Pack Secondary
packaging
system
NO
Redesign package or
reformulate
Setting-up
a Functional
Stability Unit
Each area is fundamental to the long-
I
n setting-up a stability unit it is
necessary to highlight some current term success of the firms products,
deficiencies found in Pharmaceutical whether the products are New Drugs
Stability Departments, as well as (NDAs), ANDAs or simply OTCs.
indicating the necessary control Departments Impacted
structures required for the efficient The stability department(s) must service
operation of a functional Stability the Development Department (or
Department. The structure of a practical R&D), the Regulatory Batches (those
and operational proven stability submitted to the authorities) and the
department is herewith described. Production Department (where each
Stability Control. commercial product is placed on
Stability control is achieved through stability once a year - i.e. only one
standard operational systems - namely batch of each strength and largest pack
proper stability documentation, size).
sufficient control SOPs and acceptable The Stability Requirements
monitoring equipment and laboratory The main stability operations are:
facilities. ⇒ Development Stability
The analytical testing section • Stability testing during the key
(personnel and equipment) must be of product development stages (i.e.
sufficient size to adequately perform the stability testing prior to the pivotal
stability tests in the required time. batch used for regulatory filing).
Stability testing depends on good ⇒ Regulatory Stability
timing. Consistently late drug product Stability testing of ANDA / AADA FDA
testing is of little scientific or regulatory filed batch(es):-
value. ♦ Original Generic Applications
Number of SOPs required submitted to FDA.
Stability SOPs number about 45 to 50 ♦ Amended Applications (before file
for a well managed and organized approval.)
stability department to operate ♦ Supplementary Applications
efficiently within current GMP. (changes after approval.)
A comprehensive list of the stability
⇒ Production Stability
control SOPs and some SOP
Stability testing - annually on a
summaries, controlling key functions
representative full production batch.
are included in this issue to highlight
the many operational details required. ♦ One production batch per product,
In Generic and Researched-based per strength, per year.
analytical laboratories, stability testing ♦ Annual Reports - ongoing stability
is performed in three target areas. commitments per filed application.
Do - train and re-train staff in the Don’t - allow the stability room to be
correct use and understanding of used as a stability office, where
current SOPs. personnel are continually entering and
Do - check the firms SOPs adequately leaving the controlled facility.
cover all aspects of stability operations Don’t - allow an air-conditioned 22o -
required by the FDA or Agency. 25oC stability office to function as a
Do - insure the instructions and details 25oC climatic room.
in the SOPs are adequate and sufficient
Don’t - store the 25oC long term
to assure consistent and repeated
stability samples in an office.
operation by staff, reading the SOPs.
(In terms of GMP compliance such a
Do - check staff are aware of latest facility is inadequate and the
edition of the Stability SOPs, affecting environment cannot be controlled).
their day-to-day work.
Don’t - install unreadable chart
Do - provide frequent departmental temperature recorders due to the
training in ‘reviewing and under- smallness of the rotating chart.
standing’ the principles of the SOPs. (Out-of-specifications temperatures are
not adequately shown on these charts,
ALWAYS KEEP as the range divisions on the chart are
DEPT. SOPs cramped and often too small. Narrow
ON SITE chart sensitivity scales are generally
( E l e c t r o n i c a l l y unsuitable and unreadable. The
I f p o s s i b l e ) compliance value of such a temperature
recording system is of minimal value
Do - insure operational personnel are and open to agency challenge).
aware of the latest editions of the SOPs Do - insist that current recording
and where they can be located in their devices are fitted with larger chart
stability department (All SOPs on Site). recorder so that the daily temperatures
Do - insure they are able to refer to the and OOS values can be read with
SOPs for rapid guidance in performing accuracy and precision.
their routine daily duties and tasks. Do - insure there is a system for 60%
Do - insure supervisors and personnel RH control (environmental humidity).
have signed a ‘Read and Understood’ Do - insure the stability room has
form annually indicating full awareness sufficient temperature probes at the
of the SOP contents. upper and lower levels of the room
Do - insure SOP distribution is where the stability samples are being
adequate and the SOP Change Control stored.
System really works and is consistently Do - construct a dedicated stability
on time. room with controlled environmental
Do - insure the 25oC climatic area for facilities that maintain the temperature
storing the ANDA / NDA and OTC at 25o C (± 2o C) and the relative
stability samples at 25o C (± ± 2o) is a humidity at 60% RH (± 5%).
controlled environment room. Do - install the 30o and 40o C climatic
Do - insure access is through an chamber units inside the controlled
controlled-access door, that does not stability areas or rooms.
affect the environmental temperature - Don’t - allow stability samples for
every time the door is opened.
ANDA/NDA and OTC (development, or
Do
ØPAI×
- insure that SOPs are readily
available and routinely followed and
updated (i.e. after a change or
annually). OBSERVATIONS
on
(The lack of a full set of stability SOPs
and the fact that the SOPs are
Stability
incomplete or that stability personnel
are poorly trained on the contents of the
SOPs is strong evidence to an agency
that the firm’s stability testing program ♦ Traceability of retested stability
is not in current GMP compliance). samples difficult and inconsistent.
Do - insure that it is not possible, for a
sample in a stability program to remain ♦ Traceably of raw data inconsistent.
untested after the ‘due date’ and thus
skip the designated ‘testing interval’. ♦ No written procedures for reporting
stability results precisely.
Do - insure the Certificate of Analyses
are not out of date for time zero when ♦ ‘Corrected’data substituted on FDA
the sample is eventually placed on summary data sheets.
stability at a ‘start date’ several months
after the initial C. of A. was performed. ♦ Use of ‘white-out liquid’ in stability
[Reason - the sample assay value reports to obscure test results.
potency may have degraded by several
months aging which would not be ♦ Annual reports to FDA not accurate
reflected by the initial certificate of or authentic.
analysis - some time earlier].
♦ Lack of stability and analytical SOPs
Insure to insure GMP compliance.
all Avoid
C of A's any
are White-0uts
in-date ♦ Stability data reports not internally
audited and reviewed.
Do - insure the presence of stability
SOPs controlling the maximum time ♦ Data transfer from raw documents to
period [30 days] between initial testing final report not verified.
(Certificate of Analysis at time zero)
and the ‘Start Date’ of the stability study ♦ No review of temperature/RH charts.
in order not to invalidate the initial
stability results. ♦ Uncontrolled storage of charts makes
retrospective temperature/RH chart
Do - insure that all the stability SOPs review, difficult and time consuming.
are updated according to the firm’s
No written emergency procedures
SOP index.
after equipment breakdowns.
3
Stability
SOP
Development
‘…operating a functional stability unit…
T
his section summarizes the Handling the standard procedures
Stability Units’ foremost Standard correctly may well establish the validity
Operating Procedures (SOPs). or non-validity of the firms stability
Handling SOPs in an ordered manner programs and the actual stability
may well be the solution to the effective results obtained.
development of a generic or innovative The following SOP summaries,
drug development program, not only to represent a minimum number of
place the newly formulated drug essential stability study SOPs required
product on the fast track to approval but to maintain an operational stability
hopefully to save embarrassing department for either a generic or
moments during a pre-approval innovative (researched-based) drug
inspection (PAI) should the agency development program and in full GMP
investigator stumble onto failing drug compliance for the stability testing of
product stability results in a product- developmental, regulatory and once a
specific PAI review. year commercial production batch lots.
Key Standard Operating Procedures 4
are summarized to highlight the myriad S-005-02-01YY Indexing
of procedures required for the correct procedure for Stability Studies.
handling of stability results and stability
The purpose of this standard operating
failures in an ongoing drug stability
procedure is to establish an index and
study, - be it a developmental or a final
an annual supplementary index for
formula i.e. a finished product ready to
stability study SOPs. The
go for submission.
supplementary index allows for new
How does your firm shape up in this SOPs, or updated existing SOPs, to be
stability line-up? If you don’t have the indexed in the supplement and
Stability SOP in place, - what is the firm distributed in real time.
doing about it? How is the stability 4
department handling the specific S-010-02-01YY Index for
stability requirements? Have all
Stability Studies.
stability programs and protocols
The purpose of this standard operating
involving the following subject matters
procedure is to index the Stability SOPs
been thoroughly aired and discussed
as shown above. 4
in your firms stability unit ?
1. PURPOSE
The purpose of this Standard Operating Procedure is to document an audit check list for
departmental training purposes of common practice to follow and pitfalls to avoid when
performing stability studies.
2. RESPONSIBILITY
ˆ Symbol indicates work is performed by Stability Manager.
‰ Symbol indicates work is performed by Stability Technicians.
3. FREQUENCY
Performed in the stability department.
4. PROCEDURE
In setting-up a stability unit it is necessary to highlight common deficiencies found in
Pharmaceutical Stability Departments, as well as indicating the necessary control structures
required for the efficient operation of a functional Stability Department.
The structure of a practical and operational proven stability department is emphasized with:
• correctly formatted Stability Reports (for agency review chemists).
• adequate environmental control on temperature and humidity (review of recording
graphs) - reviewed by PAI site inspectors.
• skillfully written SOPs - for efficient daily operation (reviewed during PAI site visits).
Do - insure SOP distribution is adequate and the SOPs Change Control System really
works - and on time.
Do - monitor and approve proposed changes to Stability SOPs.
Do - insure the 25 oC climatic area for storing the ANDA and OTC stability samples at
25oC (± 2o) is a controlled environment room.
Do - insure access is through an controlled-access door, that does not affect the
environmental temperature - every time the door is opened.
Don’t - allow the stability room to be used as a stability office, where personnel are
continually entering and leaving the controlled facility.
Don’t - allow an air-conditioned 22o -25o C stability office to function as a 25o C climatic
room.
Don’t - store the 25o C long term stability samples in an office.
(In terms of GMP compliance such a facility is inadequate and the environment cannot be
controlled).
Don’t - install unreadable chart temperature recorders due to the smallness of the rotating
chart. (Reason: out-of-specifications temperatures are not adequately shown on the charts, as the
range divisions on the chart are cramped and often too small. Narrow chart sensitivity scales are
generally unsuitable and unreadable. The compliance value of such a temperature recording system
is of minimal value and open to agency challenge).
Do - insist that current recording devices are fitted with larger chart recorder so that the
daily temperatures and OOS values can be read with accuracy and precision.
Do - insure there is a system for 60% RH control (environmental humidity).
Do - insure the stability room has sufficient temperature probes at the upper and lower
levels of the room where the stability samples are being stored.
Do - construct a dedicated stability room with controlled environmental facilities that
maintain the temperature at 25o C (± 2o C) and the relative humidity at 60 % RH (± 5%).
Do - install the 30o and 40o C climatic chamber units inside the controlled stability areas or
rooms.
Don’t - allow stability samples for ANDA and OTC (development, or production samples) to
be stored in cardboard boxes on cramped shelving (i.e. stacked one on top of the other.
Drug Products need to be properly exposed to the controlled environment - this requires
orderly storage on appropriate and spacious shelving. Products may not be stored
indiscriminately in cardboard boxes). The samples are not exposed to the environment
uniformly as they are protected by the insulating cardboard boxes in which they are stored.
Thus the lower samples are screened by the newer samples and a uniform controlled
exposure to temperature and humidity is not generally achieved.
The older stability samples at the bottom of the cardboard box will be temperature and
humidity screened by the several upper sample layers.
Do - avoid product exposure to large seasonal variations which do not keep the
temperature in (non-insulated) stability rooms within a 2o C range of 25o C, in either
winter or summer.
Do - avoid uneven room temperature exposures (near doorways, vents, fans.)
Do - insure the samples are arranged on the shelving in a neat, orderly manner.
Do - insure there is not a large across room-variation in temperature and humidity. Both
these variables must be adequately controlled (< 5%).
Do - insure the upper and lower shelves have been challenged for temperature
compliance. (A single chart recorder probe does not record the temperature accurately at
which all the stability samples are stored. Multiple probes are necessary - i.e. > 2 upper and
2 lower.
Do - insure the room temperature validation studies have been conducted to insure the
firm is aware of the actual storage parameters of the stability ANDA and OTC test samples.
Do - insure there is a substantive review and control of stability temperature recorders or
charts.
Do - insure temperature / RH charts are reviewed for out-of-specification (OOS)
temperature and RH values.
Do - insure the stability room charts are adequately signed and filed in an rapid retrieval
system.
Do - insure adequate quality assurance evaluation is performed on the recording charts.
Do - insure there is corrective action taken when the stability temperature goes out of the
specifications (OOS).
Do - insure that is possible for the firm to conclusively assure the FDA that the filed
ANDAs were held at 25o C, 40o C (±2o C) for the required storage periods of 3, 6, 9, 12,
18, 24, 36, etc. months.
Do - insure a corrective action SOP exists - to determine the procedures to follow after a
failure of the recording equipment or power supply during an ongoing stability study.
Do - insure corrective actions are carried out, documented and closed.
Do - insure there are written emergency procedures for the use of calibrated hand-
thermometers and recording logbooks due to recorder or stability probe failures.
Do - insure there is no lost data and full traceability of stability test points.
Do - insure summary data sheets containing ‘failed analysis results’ are meticulously
signed and filed.
Do - insure there exists a well documented reporting system for the repeat testing of
stability data, according to written SOPs.
Do - insure traceability of ALL tests performed via the laboratory work-sheets, resulting in
full credibility of the laboratory test results.
Do - investigate thoroughly if it appears that the stability data is tested and repeat tested
until it passes.
Do - insure established procedures for investigating abnormal assay fluctuations or out-of-
specification (OOS) results in the analytical and microbial stability testing program, is both
operational and functional.
Do - insure OOS SOPs are written and the principles of the Judge Wolin’s decisions are
followed and properly investigated.
Do - review and audit stability documentation in order to establish the authenticity of the
stability test results reported to the FDA in ANDAs, Supplements or Annual Reports. Insure
there is a formal pre-submission internal auditing program .
Do - insure the firms does verify the transfer of raw data values from the laboratory
workbooks to the final computer stability print-out reports.
(Where intermediate summary sheets and analysis request forms are used, these
intermediate data sheets should be signed and stamped as bona fide and accurate by
Quality Assurance).
Do - insure the final stability study is signed off by the Director of Quality Control and the
firm has a SOP specifying the acceptance and sign-off procedure for a completed stability
study, to ensure that the study is complete and accurate.
Do - insure that no laboratory raw data is unavailable or missing in support of the Stability
Summary Data Reports.
Do - insure proper cross-referencing of laboratory notebooks and worksheets with
computerized documentation prior to data being submitted to the FDA.
Do - insure retrospective audits trails of ANDA stability reports to summary data sheets and
back to laboratory workbooks clarify that the FDA filed data can be supported by the raw
laboratory test data.
Do - insure the firm does have a comprehensive and functional laboratory data reporting
system for test results.
Do - insure that data points are not missing (e.g. pH values; missing potency from crimp-
end of semi solid tubes etc.).
Do - insure stability test values are not different from the filed values.
Do - insure the use of bound and numbered laboratory notebooks.
Note - The use of unnumbered analytical worksheets for recording analytical data should be
discontinued and is not in GMP compliance.
Do - insure that stability data is not selectively screened prior to computerization.
Do - insure the absence of discrepancies and different values in ANDA Annual Reports
and the original laboratory raw data.
[Case study:- Review of the annual report prepared for the FDA showed that the ongoing
stability testing as per ANDA commitment showed an original report in the stability files with
a test data line covered with “white tape”. This data report was photocopied and sent to
the FDA. The photocopy did not reveal the ‘white-out’ data in question.]
Do - insure traceability of workbook reference page numbers and dates relating to the
original raw data in laboratory workbooks.
Do - insure the traceability of any repeat testing performed on the stability samples is
clearly referenced on the stability documentation used to prepare the computerized stability
reports.
Do - insure the need to prepare an SOP for cross-referencing laboratory notebook data
with computerized stability test result documentation.
Do - insure all repeating testing performed at the same test interval must be cross-
referenced - all together.
Note: a reviewer requires to audit all testing performed on the stability test sample and not
only the raw data in the laboratory notebooks that have passed the stability check
specifications.
Do - insure all stability data points are present and are in full compliance with the pre-
written stability protocol.
Do - insure a full review of the stability protocol and a comparison of the test procedures
carried out on the stability samples - at each test station - to highlight any incidence where
stability data points may be absent or OOS.
Do - insure that no raw data is omitted from the stability reports or in the Annual Reports
submitted to the FDA.
Do - insure stability SOPs are adequate and routinely reviewed for GMP compliance by
written in-house audits.
Do - insure the existing SOPs do control the functions of the stability department. (45-50
Stability SOPs are a minimum prerequisite to operate a stability department for an
innovative or generic drug manufacturing company).
Do - insure is that SOPs are not deficient both in the content and detail.
The lack of suitable SOPs in a stability department may result that much of the stability management
and testing of the stability samples as erratic and out-of-control - resulting in a failed PAI review.
Do - insure that SOPs are readily available and routinely followed and updated (i.e. after a
change or annually - The lack of a full set of stability SOPs and the fact that the SOPs are
incomplete or that stability personnel are poorly trained on the contents of the SOPs is
strong evidence to an agency that the firm’s stability testing program is not in current GMP
compliance).
Do - insure samples are analyzed on- time using; First-In-First-Out (FIFO).
Do - insure that it is not possible, for a sample in a stability program to remain untested
after the ‘due date’ and thus skip the designated ‘testing interval’.
Do - insure the Certificate of Analyses are not out of date for time zero when the sample is
eventually placed on stability at a ‘start date’ several months after the initial C. of A. was
performed.
[Reason - the sample assay value potency may have degraded by several months aging which would
not be reflected by the initial certificate of analysis - some time earlier].
Do - insure the presence of stability SOPs controlling the maximum time period [30 days]
between initial testing (Certificate of Analysis at time zero) and the ‘Start Date’ of the
stability study in order not to invalidate the initial stability results.
Do - insure that all the stability SOPs are updated according to the firm’s SOP index.
3
•‚ƒ„…†‡Œ•Ž
[End of Document]
Development
SOPs
‘…the essential internal standard system of a
successful drug development unit…'
INDEX OF
PHARMACEUTIC
PHARMACEUTICAL
AL
STANDARD
OPERATING
PROCEDURES
Drug Development
S tandard
O perating
P rocedures
T
his year 2000 SOP INDEX A drug researcher must keeps a
SUMMARY is intended for record of every detail of the product
individuals or groups development - both the advances
responsible for the and the failures of the experimental
management and operation of the batch lots. SOPs also demonstrate
generic drug development units. It is that you are following a key rule of
divided into four sections, a good researcher: The research
pharmaceutical, analytical, procedures must be fully described
microbiological and stability and in order that the methods can be
equally valuable for the operation duplicated and replicated as
and control of the CMC (chemistry, necessary by various unit personnel.
manufacturing and control) section
of a NDA researched-based unit. The Standard Operation
Procedures chosen fully represent a
All pharmaceutical companies cross-section of the SOPs needed
conducting drug research and for a drug development unit to
development must have SOPs. The operate efficiently and to produce
primary purpose of the SOP is to drug products on time.
translate the various regulations and
guidelines, which are open to The updated index supplies all the
interpretation, into clear and major procedures required, while
concise sets of instructions. the selected 45 summary SOPs
describe the purpose and the
Essentially generic development principles generally needed to meet
can be distilled into standard the scientific, regulatory and at
development procedures which any times GMP objectives of a well run
good drug developer would apply. A stability unit.
researcher conducts work according
to a documented set of procedures - Carefully written and structured
which hopefully represent the best SOPs will save research-based
and most current methods available firms and generic developers both
i.e. drug development using “state- time and development dollars.
of-the-art” techniques.
INDEX OF
PHARM A C E U T I C A L D E V E L O P M E N T
SOPS
SOP CONTROL
P-000-01-2000 Template for Pharmaceutical Development SOPs.
P-005-01-2000 Indexing procedure for Pharmaceutical Development SOPs.
P-010-01-2000 Index for Pharmaceutical Development SOPs.
P-015-01-2000 Signing procedures for Pharmaceutical Development SOPs.
P-020-01-2000 Numbering and format of Pharmaceutical Development SOPs.
P-025-01-2000 Circulation of Pharmaceutical Development SOPs.
P-030-01-2000 Annual Review of Pharmaceutical Development SOPs.
DEVELOPMENT NOTEBOOKS
P-035-01-2000 Issue and use of pharmaceutical development notebooks
P-040-01-2000 Signing procedures for development notebooks
P-045-01-2000 Recording pre-formulation and development formula in
development notebooks.
P-050-01-2000 Recording manufacturing instruction in development notebooks
P-055-01-2000 Recording IPQC specifications in development notebooks
P-060-01-2000 Recording finished product specifications in development
notebooks.
P-065-01-2000 Review & auditing of pharmaceutical development notebooks
P-070-01-2000 Correction procedures in development notebooks &
documentation
P-075-01-2000 Archiving of development notebooks.
DEVELOPMENT FORMULA
P-085-01-2000 Operating procedures for product development.
P-090-01-2000 Formulation of ANDA topical preparations
P-095-01-2000 Formulation of ANDAs to Q1Q2 Status (semisolids)
P-100-01-2000 Validation requirements for Product Development
P-105-01-2000 Vendor Certification requirements for Product Development
P-110-01-2000 Check list for a pharmaceutical Development Report
P-115-01-2000 SOP for Development Reports
OF INDEX
PHARMACEUTICAL DEVELOPMENT SOPS
DEVELOPMENT FORMULA
P-120-01-2000 Formulation of CR / ER ANDA Oral Tablet Preparations
P-125-01-2000 Establishing an IVIVC in Extended Release Oral Dosage Forms
P-130-01-2000 Establishing a level A IN-VITRO IN-VIVO correlation
P-135-01-2000 Establishing a level B IN-VITRO IN-VIVO correlation
P-140-01-2000 Establishing a level C IN-VITRO IN-VIVO correlation
P-145-01-2000 Establishing a level A IN-VITRO IN-VIVO correlation
P-150-01-2000 Evaluating the predictability of a level A - IVIV Correlation
P-155-01-2000 Development and Evaluation of a level C IVIV Correlation
DEVELOPMENT REPORTS
P-160-01-2000 List of FDA Guidance documents impacting on product IR and
CR development dosage forms.
P-165-01-2000 Setting up a general Development SOPs.
P-170-01-2000 Standard Procedures for Generic Product Development
P-175-01-2000 Setting up a Product Specific Development SOPs.
P-180-01-2000 Product Specific Development SOPs for CR Tablets - Contents.
P-185-01-2000 Setting up a Product Specific ER Development SOP.
P-190-01-2000 Setting up IVIVC for Extended Release Oral Dosage Forms
P-195-01-2000 Contents of a Development SOP - ER Oral Tablets.
Active materials
P-200-01-2000 Active Drug Substances for Generic Drugs
P-205-01-2000 Developing Product Formula with approved Actives
P-210-01-2000 R&D Inventory Records for the Active Drug Substance
P-215-01-2000 Vendor Certification Requirements for Approved Actives.
P-230-01-2000 Decision tree for establishing impurity acceptance criteria.
P-235-01-2000 Decision tree for establishing degradation acceptance criteria
P-240-01-2000 Decision tree for establishing particle size acceptance criteria
P-245-01-2000 Decision tree for establishing polymorphism existence
P-250-01-2000 Decision tree for establishing microbiological testing
P-255-01-2000 Decision tree for evaluating chiral actives
OF INDEX
PHARMACEUTICAL DEVELOPMENT SOPS
Non-Active materials
P-285-01-2000 Non-active ingredients for ANDA formula development
P-290-01-2000 Use of Purified Water USP in Product Development
P-295-01-2000 Checking excipients in the FDA ‘Inactive Ingredient Guide’
P-300-01-2000 Evaluation and Requirements of Release Controlling Excipients
P-305-01-2000 Justification and functionality of the Release Controlling
Excipient
Container-Liner-Closure systems
P-310-01-2000 Container-Liner-Closure systems for Generic Development
P-315-01-2000 Documentation requirements for Container/Closure systems
P-320-01-2000 Check list for Container-Liner-Closure Documents
In-process controls
P-325-01-2000 Choice of IPQC limits.
P-330-01-2000 Qualification of IPQC limits.
P-335-01-2000 Qualification of manufacturing process specification limits.
P-340-01-2000 In process control on bulk products
P-345-01-2000 Time limitations on manufacturing processing stages
Contract laboratories
OF INDEX
PHARMACEUTICAL DEVELOPMENT SOPS
SOP Number Development Study Procedure
Pivotal Batch
P-420-01-2000 Pivotal Batch requirements
P-425-01-2000 In-process sampling & testing procedures of tablets, caplets
and capsules for pivotal batches
P-430-01-2000 Do’s and Don’ts when preparing for pivotal batches
P-435-01-2000 Check list for Pivotal Batch Documentation
P-440-01-2000 Side by side comparison for Pivotal and Validation Batch
OF INDEX
PHARMACEUTICAL DEVELOPMENT SOPS
SOP Number Development Study Procedure
Biostudy
P-445-01-2000 Do’s and Don’ts when preparing for pivotal Biostudies
P-450-01-2000 Dissolution requirements for Biostudies
P-455-01-2000 Dissolution Testing for Solid Oral Dosage Forms
P-460-01-2000 Dissolution Testing for Suspended Oral Dosage Forms
P-465-01-2000 Check List & Documentation for and IVIVC/Pilot Study
P-470-01-2000 Check List for Biostudy Documentation
Sanitation
P-475-01-2000 Good House Keeping Practice in a Small Scale Development
Unit
P-480-01-2000 Cleaning and Sanitation Procedures for Small Scale
Development Unit
P-485-01-2000 Validation of Cleaning procedures for Small Scale
Manufacturing Equipment
P-490-01-2000 Garmenting procedures for development personnel
Chart Control
P-495-01-2000 Routine signing and checking of temperature charts
P-500-01-2000 Review & control of temperature & humidity recording charts
OF INDEX
PHARMACEUTICAL DEVELOPMENT SOPS
SOP Number Development Study Procedure
Contract laboratories
P-555-01-2000 Auditing procedures for a contract laboratory.
P-560-01-2000 Mail / fax auditing procedures for a contract laboratory.
Reviewing documentation
P-610-01-2000 Review And Auditing Development Documentation.
P-615-01-2000 Review And Auditing The Process Qualification Batch
Documentation.
P-620-01-2000 Review And Auditing The Pivotal Batch Documentation.
Closing a study
P-625-01-2000 Accepting and signing-off a completed development study.
INDEX
OF
ANALYTICAL
STANDARD
OPERATING
PROCEDURES
Drug Development
INDEX OF
ANA L Y T I C A L D E V E L O P M E N T
SOPS
INDEX No SOPs
A-001-01-2000 Indexing procedure for analytical SOPs.
A-010-01-2000 Index of analytical SOPs.
A-012-01-2000 Authorization signatures for analytical SOPs.
A-015-01-2000 Numbering and format of analytical SOPs.
A-020-01-2000 Circulation of analytical SOPs.
A-025-01-2000 Annual Review of analytical SOPs.
A-030-01-2000 Reserved.
Development Notebooks
A-035-01-2000 Issue and use of analytical development notebooks
A-040-01-2000 Signing procedures for analytical notebooks
A-045-01-2000 Entering raw data in laboratory notebooks
A-050-01-2000 Using USP terminology in analytical methods
A-055-01-2000 Verifying analytical calculations performed by (in-house)
computer programs
A-060-01-2000 Release of Results from the Analytical R&D Laboratories.
A-060-02-2000 Reviewer Checklist .
Auditing
A-065-01-2000 Review and auditing of analytical laboratory notebooks
A-070-01-2000 Correction procedures in laboratory notebooks
A-075-01-2000 Archiving of laboratory notebooks
A-080-01-2000 Laboratory Note Book Checklist.
Incoming samples
A-095-01-2000 General Analytical Sample Preparation.
A-100-01-2000 Receipt and logging-in of analytical laboratory samples
A-105-01-2000 Storage of samples prior to testing
A-110-01-2000 Storage time limits of samples prior to testing.
A-115-01-2000 Disposition of tested laboratory samples (including time limits).
A-120-01-2000 Reserved.
INDEX OF
AN A L Y T I C A L D E V E L O P M E N T
SOPS
INDEX No SOPs
Reagent and solutions
A-125-01-2000 Handling and preparation of analytical standards
A-130-01-2000 Handling and preparation of volumetric solutions
A-135-01-2000 Labeling requirements of reagents and solutions
A-140-01-2000 Preparation and storage of analytical glassware.
A-145-01-2000 Reserved.
Test methods
A-150-01-2000 Availability and control of approved test methods.
A-155-01-2000 Updating Pharmacopeial methods with supplemental
monographs.
A-160-01-2000 Abbreviated Raw Materials testing Procedures.
A-165-01-2000 Approval signatures for Raw materials and Approved suppliers.
A-170-01-2000 Retesting Procedures.
Calculations
A-175-01-2000 Recording and checking of method calculations
A-180-01-2000 Procedures for rounding off analytical numbers
Active materials
A-190-01-2000 Active Drug Substances for Generic Drugs
A-195-01-2000 Developing Product Formula with approved Actives
A-200-01-2000 Development Inventory Records for the Active Drug Substance
A-205-01-2000 Reserved.
Drug substance
A-210-01-2000 Drug substance impurity assays
A-215-01-2000 Drug substance impurities profiles
A-220-01-2000 Drug substance specifications
A-225-01-2000 Drug substance approval procedures
A-235-01-2000 Drug substance approved suppliers
INDEX OF
AN A L Y T I C A L D E V E L O P M E N T
SOPS
INDEX No SOP
Drug Product
A-250-01-2000 Drug substance impurity assays
A-255-01-2000 Drug substance impurities profiles
A-260-01-2000 Drug substance Specifications
A-265-01-2000 Limit test on impurities
A-270-01-2000 Validation of limit tests for impurities
A-272-01-2000 Validation of Assay and/or Impurities Determination
A-275-01-2000 Assay determination by HPLC and GC methods.
A-276-02-2000 Assay determination by HPLC and GC methods -Details.
Container-liner-closure systems
A-280-01-2000 Testing Container-Liner-Closure systems for Generic
Development
Sample preparation
A-290-01-2000 General analytical sample preparation
A-295-01-2000 Number of samples and injections for assays
A-300-01-2000 Standards and system suitability for HPLC testing
A-304-01-2000 Working and Impurity Standards - Use and Qualification
A-305-01-2000 Working with Reference Standards and In-house Standards.
Validation
A-310-01-2000 Using ID numbers for identifying laboratory instrumentation.
A-315-01-2000 Validation of stability-indicating (S-I) methods
A-320-01-2000 Validation of in-house analytical methods
A-325-01-2000 Using stability indicating (S-I) methods
A-335-01-2000 Analytical methods not requiring (full) validation
A-340-01-2000 Contents of an analytical validation protocol
A-345-01-2000 Standardizing and transferring S-I methods and assay
validations.
A-350-01-2000 Change Control Procedures.
Contract laboratories
A-355-01-2000 Auditing procedures for a contract analytical laboratory.
A-365-01-2000 Mail/fax auditing procedures for a contract laboratory.
INDEX OF
AN A L Y T I C A L D E V E L O P M E N T
SOPS
INDEX OF
AN A L Y T I C A L D E V E L O P M E N T
SOPS
INDEX OF
AN A L Y T I C A L D E V E L O P M E N T
SOPS
SOP Number ANALYTICAL STUDY PROCEDURE
Reviewing documentation
A-650-01-2000 Review and Auditing analytical data.
A-655-01-2000 Auditing the Process Qualification Batch analytical data.
A-660-01-2000 Review and Auditing the Pivotal Batch analytical data.
A-665-01-2000 Review and Auditing Stability Batch analytical data.
Closing a study
A-670-01-2000 Accepting and signing-off a completed analytical study.
INDEX OF
MICROBIOLOGICAL
STANDARD
OPERATING
PROCEDURES
Drug Development
INDEX OF
MICROBIOLOGY DEVELOPMENT
SOPS
SOP Control
M-005-01-2000 Indexing procedure for Microbiology SOPs.
M-010-01-2000 Index for Microbiology SOPs.
Notebooks
M-015-01-2000 Issue, use, and disposition of microbiological laboratory
notebooks
Samples and Sampling
M-020-01-2000 The use of sterile sampling containers.
M-025-01-2000 Representative sampling procedures.
M-030-01-2000 Labeling of sample containers.
M-035-01-2000 Receipt and logging of laboratory samples.
M-040-01-2000 Storage of samples before and after testing.
M-045-01-2000 Storing the Microbiology study samples under refrigerated
conditions prior to analysis.
M-050-01-2000 Number of samples required for performing microbiology tests.
M-055-01-2000 Storage time limitations of samples prior to testing
Media
M-075-01-2000 Labeling and expiration dating of prepared media
M-080-01-2000 Disposition of microbiological media and samples
M-085-01-2000 Preparation, storage and use of microbiological media.
INDEX OF
MICROBIOLOGY DEVELOPMENT
SOPS
In-process controls
M-115-01-2000 Representative sample procedures on bulk products
M-120-01-2000 In process control on bulk products
M-125-01-2000 Time limitations on bulk product in process controls
Laboratory House-keeping
M-155-01-2000 Procedures for reduction of bioburden in the microbiological
laboratory
M-160-01-2000 The use and rotation of disinfectant swabbing solutions.
M-165-01-2000 Prevention of contamination of media plates
M-170-01-2000 Preparation, sterilization and storage of laboratory glassware
and equipment
Culture control
M-175-01-2000 Procedures for receipt, storage and handling of ATCC cultures
M-180-01-2000 Handling Certificate of Analysis for ATCC cultures
M-185-01-2000 Limitation on transfer procedures for ATCC cultures
INDEX OF
MICROBIOLOGY DEVELOPMENT
SOPS
Formula Control
M-210-01-2000 Recording and checking of method calculations
M-215-01-2000 Procedures for rounding-off recorded numbers
Investigation reports
M-220-01-2000 Procedures for handling abnormal or OOS results in a
microbiology study.
M-220-01-2000 Procedures for repeat testing
M-230-01-2000 Investigation reports after repeat testing
M-230-01-2000 Procedures for invalidating test results
Aseptic practice
M-240-01-2000 Periodic monitoring of lamina flow units
M-240-01-2000 Aseptic working practice and techniques for laminar flow units
Environmental monitoring
M-250-01-2000 Bioburden mapping of laboratory environment
M-250-01-2000 Bioburden mapping of manufacturing environment
M-260-01-2000 Bioburden evaluation of manufacturing equipment
M-260-01-2000 Bioburden sampling and evaluation of the environment air
M-270-01-2000 The operation and use of Biotest Hycon air RCS sampler
M-270-01-2000 Bioburden evaluation of personnel hands and clothing
M-280-01-2000 Swabbing procedures for surface evaluation.
Chart Control
M-290-01-2000 Routine signing and checking of temperature charts
M-290-01-2000 Review and control of temperature and humidity recording
charts.
INDEX OF
MICROBIOLOGY DEVELOPMENT
SOPS
Sanitation
M-335-01-2000 Sanitation and housekeeping requirements of incubators.
M-340-01-2000 Good House Keeping practice in a microbiological laboratory
M-345-01-2000 Cleaning and sanitation procedures for incubators and
refrigerators
M-350-01-2000 Garmenting procedures for microbiological personnel
Contract laboratories
M-380-01-2000 Auditing procedures for a contract laboratory.
M-385-01-2000 Mail/fax auditing procedures for a contract laboratory.
Development SOP
M-390-01-2000 Microbiology development procedures for new products.
Reviewing documentation
M-410-01-2000 Review and auditing microbiology documentation.
M-415-01-2000 Reporting the test results of a microbiology study.
Closing a study
M-420-01-2000 Accepting and signing-off a completed microbiology study.
INDEX
OF
STABILITY
STANDARD
OPERATING
PROCEDURES
Drug Development
INDEX OF
STABILITY
SOPS
The following index represents an adequate set of standard operating procedures for a stability
department. In order for a stability department to function efficiently the principles described in
these over +45 standard operating procedures are required to conduct a functional stability
study. SOP examples are provided as a base. All SOPs listed are not provided.
SOP CONTROL
S-001-01-2000 Format and Layout of Standard Operating Procedures
S-005-01-2000 Indexing procedure for Stability Studies.
S-010-01-2000 Index for Stability SOPs.
STARTING A STUDY
S-015-01-2000 Initiating a Stability Study.
S-020-01-2000 Contents of a Stability Protocol.
S-025-01-2000 Setting the ‘Start date’ for a Stability Study.
S-030-01-2000 Determining the ‘Due dates’ for a Stability Study protocol.
S-035-01-2000 The initial Certificate of Analysis at T o for a Stability Study.
STUDY PARAMETERS
S-040-01-2000 Setting limits for check specifications in a Stability Study.
S-045-01-2000 Number and size of batches for stability testing.
SAMPLING
S-050-01-2000 Number of samples required for performing stability tests.
S-060-01-2000 Labeling of Stability Study Samples.
S-065-01-2000 Storage configuration of samples in a stability environment.
S-070-01-2000 Storing the stability study samples under controlled conditions prior to analysis.
ACTIVE DRUG
S-075-01-2000 Stress testing the bulk drug substance for stability analysis.
STUDY CONDITIONS
S-080-01-2000 Intervals and climatic conditions for a US development Stability Study.
S-085-01-2000 Intervals and climatic conditions for a US Pivotal/Bioequivalence Stability Study.
S-090-01-2000 Intervals and climatic conditions for a US validation/PM Stability Study.
S-095-01-2000 Placing the Reference Listed Drug (RLB) on Stability.
PACKAGING PROCEDURES
S-100-01-2000 Sampling and Testing of Pivotal Batches - Tablet and Capsule Dosage Forms.
S-105-01-2000 Sampling and Testing of Pivotal Batches - Powder and Syrups for Reconstitution.
CONTAINER SYSTEMS
S-110-01-2000 Container-Liner-Closure systems for a Stability Study.
S-115-01-2000 Certification of a Container-Liner-Closure system.
TEST RESULTS
S-120-01-2000 Reporting test results of a Stability Study.
S-125-01-2000 Procedures for handling abnormal or OOS results in a Stability Study.
INDEX OF
STABILITY
SOPS
TEST METHODS
S-130-01-2000 The control of Analytical methods #’s and Edition #’s in stability documentation.
CHART CONTROL
S-150-01-2000 Recording stability study climatic conditions
S-155-01-2000 Review and control of temperature and humidity recording charts.
CORRECTIVE ACTION
S-175-01-2000 Fault correcting procedures (after breakdowns) during a Stability Study.
S-180-01-2000 Emergency procedures during a Stability Study.
IN HOUSE METHODS
S-185-01-2000 Reserved.
STOPPING A STUDY
S-190-01-2000 Conditions for stopping a Stability Study.
SELF INSPECTION
S-210-01-2000 Self inspection procedures in a stability department.
REVIEWING DOCUMENTATION
S-245-01-2000 Review and auditing stability study documentation.
S-250-01-2000 The layout and format of a regulatory stability report (a filed report)
S-255-01-2000 Documentation requirements for a Stability Study - contents of a stability dossier
CLOSING A STUDY
S-260-01-2000 Accepting and signing-off a completed stability study.
3
[End of Document]
I n t e r n a t i o n a l A s s o c i a t i o n o f G e n e r i c & I n n o v a t i v e D r u g M a n u f a c t u r e r s