Discover millions of ebooks, audiobooks, and so much more with a free trial

Only $11.99/month after trial. Cancel anytime.

The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals
The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals
The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals
Ebook642 pages6 hours

The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals

Rating: 0 out of 5 stars

()

Read preview

About this ebook

Good Manufacturing Practices (GMP) for human pharmaceuticals affects every patient taking a medicine. GMP covers all aspects of the manufacturing process, from defining manufacturing processes to systems for recall and investigation of complaints. Consumers expect that each batch of medicines they take will meet quality standards so that they will be safe and effective.

GMPs provide for systems that assure proper design, monitoring, and control of manufacturing processes and facilities. This formal system of controls at a pharmaceutical company, if adequately put into practice, helps to prevent instances of contamination, mix-ups, deviations, failures, and errors. This assures that drug products meet their quality standards.

This guidance book is meant as a resource to manufacturers of pharmaceuticals, providing up-to-date information concerning required and recommended quality system practices. It should be used as a companion to the regulations/standards themselves and texts on the specific processes and activities contained within the QMS.
As a bonus, this package contains dozens of FDA guidance documents as well as international harmonization documents (WHO, PIC/S, and ICH). A check list for GMP audit is also included based on risk management criteria. An exam complements the extra material.
LanguageEnglish
Release dateApr 30, 2014
ISBN9780873898294
The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals
Author

José (Pepe) Rodríguez-Pérez

Dr. José (Pepe) Rodríguez-Pérez is the president of Business Excellence Consulting Inc. (BEC), a Puerto Rico-based global consultant, training, and remediation firm in the areas of regulatory compliance, risk management, and regulatory training in the FDA-regulated sector. He’s also president of BEC Spain. Dr. Rodríguez-Pérez is a biologist and earned his doctoral degree in biology from the University of Granada (Spain). He served as professor and director of the Microbiology Department at one of the Puerto Rico schools of medicine, and he also served as Technical Services manager at a manufacturing plant of Abbott Laboratories in Puerto Rico. From 2003 to 2012, he was professor for graduate studies of the Polytechnic University of Puerto Rico, and he served as a Science Advisor for the FDA from 2009 to 2011. Dr. Rodríguez-Pérez is a senior member of ASQ, as well as a member of AAMI, ISPE, PDA, and RAPS. He is an ASQ-certified Six Sigma Black Belt, Quality Manager, Quality Engineer, Quality Auditor, Quality HACCP Auditor, Biomedical Auditor, and Pharmaceutical GMP Professional. He is also the author of the best-selling books CAPA for the FDA-Regulated Industry, Quality Risk Management in the FDA-Regulated Industry, The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals, Human Error Reduction in Manufacturing, and Data Integrity and Compliance, all available from ASQ Quality Press. Contact Dr. Rodríguez-Pérez at pepe.rodriguez@bec-global.com.

Related to The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals

Related ebooks

Business For You

View More

Related articles

Reviews for The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals

Rating: 0 out of 5 stars
0 ratings

0 ratings0 reviews

What did you think?

Tap to rate

Review must be at least 10 words

    Book preview

    The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals - José (Pepe) Rodríguez-Pérez

    The FDA and Worldwide Current Good Manufacturing Practices and Quality System Requirements Guidebook for Finished Pharmaceuticals

    José Rodríguez-Pérez

    ASQ Quality Press

    Milwaukee, Wisconsin

    American Society for Quality, Quality Press, Milwaukee 53203

    © 2014 by ASQ

    All rights reserved.

    Library of Congress Cataloging-in-Publication Data

    Rodríguez Pérez, José, 1961–

    The FDA and worldwide current good manufacturing practices and quality system requirements guidebook for finished pharmaceuticals / José Rodríguez-Pérez.

    pages cm

    Includes bibliographical references and index.

    ISBN 978-0-87389-869-0 (alk. paper)

    1. Pharmaceutical industry—Production standards—United States. 2. Pharmaceutical industry—United States—Quality control. 3. Drugs—Standards. I. Title.

    RS192.R63 2014

    338.476151—dc23

    2014018745

    No part of this book may be reproduced in any form or by any means, electronic, mechanical, photocopying, recording, or otherwise, without the prior written permission of the publisher.

    Acquisitions Editor: Matt Meinholz

    Managing Editor: Paul Daniel O’Mara

    Production Administrator: Randall Benson

    ASQ Mission: The American Society for Quality advances individual, organizational, and community excellence worldwide through learning, quality improvement, and knowledge exchange.

    Attention Bookstores, Wholesalers, Schools, and Corporations: ASQ Quality Press books, video, audio, and software are available at quantity discounts with bulk purchases for business, educational, or instructional use. For information, please contact ASQ Quality Press at 800-248-1946, or write to ASQ Quality Press, P.O. Box 3005, Milwaukee, WI 53201-3005.

    To place orders or to request a free copy of the ASQ Quality Press Publications Catalog, visit our website at http://www.asq.org/quality-press.

    ASQ-Logo-QPress-address-K.jpg

    This book is dedicated to all my associates at Business Excellence Consulting Inc. Thanks for your support and sharing our passion for quality.

    Preface

    Quality must be built into the product—testing alone cannot be relied on to ensure product quality.

    Good manufacturing practice (GMP) for human pharmaceuticals affects every individual who takes medicine. Consumers expect that the medicines they take will meet quality standards and will be safe and effective. Most people, however, are not aware of the existence of GMP or how government regulators ensure that drug manufacturing processes meet these basic objectives.

    The original requirements for GMP from the World Health Organization (WHO) date back to 1975. The WHO defines GMP as that part of quality assurance which ensures that products are consistently produced and controlled to the quality standards appropriate to their intended use and as required by the marketing authorization.¹ Chapter 4 discusses WHO GMP requirements.

    GMP covers all aspects of the manufacturing process: defined manufacturing process; validated critical manufacturing steps; suitable premises, storage, and transport; qualified and trained production and quality control personnel; adequate laboratory facilities; approved written procedures and instructions; records to show all steps of defined procedures were taken; full traceability of a product through batch processing records and distribution records; and systems for recall of product and investigation of complaints.

    GMP constitutes the element of quality assurance that ensures products are consistently produced and controlled to the quality standards appropriate to their intended use and as required by government authorization or product specification. GMPs are related to both production and quality control and they cannot be considered best practices; rather, they establish threshold or minimum standards that must be satisfied in order for a pharmaceutical manufacturing operation to be compliant.

    GMP provides for systems that ensure proper design, monitoring, and control of manufacturing processes and facilities. Adherence to the GMP regulations ensures the identity, strength, quality, and purity of drug products by requiring that manufacturers of medications adequately control manufacturing operations. This includes establishing strong quality management systems (QMSs), obtaining appropriate quality raw materials, establishing robust operating procedures, detecting and investigating product quality deviations, and maintaining reliable testing laboratories. This formal system of controls at a pharmaceutical company, if adequately put into practice, helps prevent instances of contamination, mix-ups, deviations, failures, and errors. This ensures that drug products meet their quality standards.

    The guiding principle of GMP is that quality is built into a product, not just tested into a finished product. Therefore, the assurance is that the product not only meets the final specifications but is made by the same procedures under the same conditions each and every time. There are many ways this is done, such as by controlling the quality of the facility and its systems, controlling the quality of the starting materials, controlling the quality of production at all stages, controlling the quality of the testing of the product, controlling the identity of materials by adequate labeling and segregation, controlling the quality of materials and products by adequate storage, and so on. All of these controls must follow prescribed, formal, and approved procedures or master formulae describing all the tasks carried out in the manufacturing and control processes.

    The GMP requirements were established to be flexible in order to allow each manufacturer to decide individually how to best implement the necessary controls by using scientifically sound design, processing methods, and testing procedures. The flexibility in these regulations allows companies to use modern technologies and innovative approaches to achieve higher quality through continual improvement. Accordingly, the c in cGMP stands for current, requiring companies to use technologies and systems that are up to date in order to comply with the regulations.

    Consumers usually cannot detect whether a drug product is safe or whether it will be effective. While cGMP requires testing, testing alone is not adequate to ensure quality. In most instances testing is done on a small sample of a batch (for example, a drug manufacturer may test a few dozen tablets from a batch that contains several million units) so that most of the batch can be used for patients rather than being destroyed by testing. Therefore, it is important that drugs are manufactured under conditions and practices established by the cGMP regulations to ensure that quality is built into the design and manufacturing process at every step. Facilities that are in good condition, equipment that is properly maintained and calibrated, employees who are qualified and fully trained, and processes that are reliable and reproducible are a few examples of how cGMP requirements help ensure the safety and efficacy of drug products.

    It is generally agreed that the cGMP requirements applicable to the manufacture of veterinary medicinal products should be very similar (if not identical) to those for the manufacture of product for human use.

    The first manufacturing and quality requirements, which later evolved into cGMPs, were issued in the 1940s in the United States by the Food and Drug Administration (FDA). In the 1969 general meeting of the WHO, the World Health Assembly issued a recommendation for the introduction of GMPs. Since then, most industrialized countries have passed laws on control procedures essential for the manufacture of drug products. The cGMP regulations originated from congressional concern that impure and otherwise adulterated drugs might escape detection under a system predicated only on seizure of drugs shown to be in fact adulterated.

    In 1962, the US Congress enacted various amendments to the Federal Food, Drug, and Cosmetic (FD&C) Act of 1938 to strengthen and broaden existing laws in the drug field so as to bring about better, safer medicine and to establish a more effective system of enforcement of the drug laws.²

    Among the amendments was a section by which a drug is deemed adulterated if its packaging, processing, holding, or manufacturing fails to conform to current good manufacturing practice (cGMP) to assure that such drug meets the requirements of this chapter as to safety and has the identity and strength, and meets the quality and purity characteristics, which it purports or is represented to possess. The FDA issued its first regulations under this section in 1963, and in February 1976, it announced a proposal to revise and update the then-current GMP regulations.

    The legal authority for the FDA to impose minimum manufacturing standards is set forth in the Federal FD&C Act, 21 U.S.C. sec. 301 et seq. Section 351(a)(2)(B) of 21 U.S.C. requires manufacturers of drugs to operate in conformance with manufacturing regulations established by the FDA. The regulations are primarily contained in Title 21 of the US Code of Federal Regulations (CFR), Parts 210 and 211, and are called the cGMP regulations. Drugs that are not manufactured in accordance with cGMP requirements, including the quality control and quality process mandates, are adulterated under the FD&C Act. The knowledge and understanding of cGMP establishes the foundation for drug product safety and quality, allowing for quality to be built into the design and production of pharmaceuticals.

    GMP requirements are supported by a central objective: to create a system of programs, policies, processes, and facilities that prevent errors and defects. Senior managers in the pharmaceutical industry are responsible for the effectiveness of this system, which is known as the pharmaceutical quality system (PQS). A PQS is successful when it ensures an ongoing state of control. In a healthy PQS, managers establish a vigilant culture of quality in which timely action is taken to prevent risks to quality. Life cycle adaptations are made to address manufacturing weaknesses and continually improve systems. An effective process performance and product quality monitoring program provides early warning of emerging quality issues. Systemic solutions are implemented rather than ineffective shortcuts. An effective PQS will ultimately support stable processes and predictable supply of quality medicinal products.

    This book is divided into eight chapters. It also includes an appendix with two tabulated comparisons: the first compares US, European, Pharmaceutical Inspection Co-operation Scheme (PIC/S), Canadian, and WHO cGMPs, while the second compares US cGMPs with effective quality system elements.

    Chapter 1 serves as an introduction to cGMP, while Chapter 2 presents how the US government regulates pharmaceutical products. Details regarding regulation of nonprescription products in the United States are also discussed.

    Chapter 3 describes how pharmaceutical products are regulated worldwide, while Chapter 4 provides details of global cGMP guides including WHO and the International Conference on Harmonization (ICH) quality series guides. PIC/S voluntary cGMP guides are also discussed.

    Chapter 5 provides a very comprehensive and detailed analysis of the US requirements and guidances for finished drug product manufacturing. It compares these requirements with those from the European Union (EU)/PIC-S and the WHO. Current enforcement issues related to cGMP are discussed in the Author’s Notes sections of this chapter.

    Chapter 6 discusses the missing subparts of this regulation: corrective and preventive action, internal audit, validations, and purchasing controls.

    The titles of the next two chapters are self-explanatory: What Are Inspectors Looking For? (Chapter 7) and Quality at Risk: The Price of Noncompliance (Chapter 8). Recalls, inspection results, warning letters, consent decrees, and the debarment list are part of the hot-issue regulatory topics covered.

    The companion CD included with the print version of this book contains cGMP regulations for sterile products produced by aseptic processing; it also includes updated data of statistical enforcement by the FDA, both domestically and abroad; a detailed glossary; and dozens of FDA guidance documents as well as international regulations (EU and Canada) and harmonization documents (WHO, PIC/S, and ICH). A very comprehensive checklist for a cGMP audit that is based on risk management criteria is also included. Finally, a comprehensive GMP exam is also included. To request these files, please email authors@asq.org.

    Notes

    WHO Expert Committee on Specifications for Pharmaceutical Preparations, Good Manufacturing Practices for Pharmaceutical Products, Technical Report Series No. 823 Annex 1 (Geneva: WHO, 1992).

    H. R. Rep. No. 2464, 87th Cong., 2d Sess. 2 (1962). See also 1962 US Cong. and Admin. News, p. 2884.

    List of Acronyms

    Table of Contents

    Preface

    List of Acronyms

    Chapter 1

    Introduction

    Facts about cGMP

    cGMP and the Quality System

    Notes

    Chapter 2

    US Current Good Manufacturing Practice

    cGMP and the FDA

    Drugs Regulated by CDER

    Foreign Manufacturers

    Notes

    Chapter 3

    International Good Manufacturing Practice

    The EU

    Canada

    Japan

    Brazil

    Australia

    China

    India

    Korea

    South Africa

    Russia

    New Zealand

    Notes

    Chapter 4

    Global Good Manufacturing Practice Guides and Harmonization

    The WHO

    International Conference on Harmonization

    Pharmaceutical Inspection Convention and the Pharmaceutical Inspection Co-operation Scheme

    Notes

    Chapter 5

    Detailed Analysis of the Requirements and Guidances

    Part 210—Current Good Manufacturing Practice in Manufacturing, Processing, Packing, or Holding of Drugs: General

    Part 211—Current Good Manufacturing Practice for Finished Pharmaceuticals

    Notes

    Chapter 6

    Missing Subparts

    Corrective and Preventive Action System

    Management Control

    Process Validations

    Notes

    Chapter 7

    What Are Inspectors Looking For?

    Types of Inspections

    Drug Manufacturing Inspections

    Preapproval Inspections

    API Inspections

    Sterile Drug Process Inspection

    Notes

    Chapter 8

    Quality at Risk: The Price of Noncompliance

    Field Alerts

    Recall Process for FDA-Regulated Products

    Form 483

    Regulatory Meetings

    Dear Health Care Provider Letters

    Untitled and Warning Letters

    Criminal Investigations

    Seizure

    Injunctions

    Consent Decrees and Disgorgements

    Debarment and Disqualification List

    Progressive Enforcement

    Notes

    Appendix

    At-a-Glance Comparisons

    Bibliography

    Useful Websites

    Chapter 1

    Introduction

    In the United States, finished pharmaceutical cGMP refers to the regulations enforced by the FDA. cGMP provides for systems that ensure proper design, monitoring, and control of manufacturing processes and facilities. Adherence to the cGMP regulations ensures the identity, strength, quality, and purity of drug products by requiring that manufacturers of medications adequately control manufacturing operations. This includes establishing strong QMSs, obtaining appropriate-quality raw materials, establishing robust operating procedures, detecting and investigating product quality deviations, and maintaining reliable testing laboratories. This formal system of controls at a pharmaceutical company, if adequately put into practice, helps prevent instances of contamination, mix-ups, deviations, failures, and errors. This ensures that drug products meet their quality standards and that patients receive safe and effective medical products.

    The cGMP requirements were established to be flexible in order to allow each manufacturer to decide individually how to best implement the necessary controls by using scientifically sound design, processing methods, and testing procedures. The flexibility in these regulations allows companies to use modern technologies and innovative approaches to achieve higher quality through continual improvement. Accordingly, the c in cGMP stands for current, requiring companies to use technologies and systems that are up to date in order to comply with the regulations. Systems and equipment that may have been top of the line to prevent contamination, mix-ups, and errors 20 or 30 years ago may be less than adequate by today’s standards.

    It is important to note that cGMPs are minimum requirements, not best practices. They establish threshold or minimum standards that must be satisfied in order for a pharmaceutical manufacturing operation to be compliant.

    The importance of cGMP lies in the fact that consumers usually cannot detect (through smell, touch, or sight) whether a drug product is safe or whether it will work. While cGMPs require testing, testing alone is not adequate to ensure quality. In most instances, testing is done on a small sample of a batch (for example, a drug manufacturer may test a few dozen units from a batch that contains several million tablets) so that most of the batch can be used for patients rather than destroyed by testing. Therefore, it is important that drugs are manufactured under conditions and practices required by the cGMP regulations to ensure that quality is built into the design and manufacturing process at every step. Facilities that are in good condition, equipment that is properly maintained and calibrated, employees who are qualified and fully trained, and processes that are reliable and reproducible are a few examples of how cGMP requirements help ensure the safety and efficacy of drug products.

    Facts about cGMP

    Government regulators (the FDA in the United States) inspect pharmaceutical manufacturing facilities worldwide using scientifically and cGMP-trained inspectors whose job is to evaluate whether the company is following the cGMP regulations. The FDA also relies on reports of potentially defective drug products from the public and the industry and often uses these reports to identify sites for which an inspection is needed. Most companies that are inspected are found to be compliant with the cGMP regulations (see companion CD for updated inspection and enforcement statistics).

    If a company is not complying with cGMP regulations, any drug it makes is considered adulterated under the law. This means that the drug was not manufactured under conditions that comply with cGMP. It does not mean that there is necessarily something wrong with the drug. The product can be perfectly safe for use, but as established in the FD&C Act, drugs that are not manufactured following cGMP requirements are adulterated.

    Consumers who are currently taking medicines from a company that was not following cGMP can be advised by the FDA not to interrupt their drug therapy, as this could have serious implications for their health. Consumers should seek advice from their health care professional before stopping or changing medications. By focusing on the procedures and processes used to make drugs, the FDA tries to ensure that the drugs meet their quality standards and are safe and effective. The impact of cGMP violations depends on the nature of the violations and on the specific drugs involved. A drug manufactured in violation of cGMP may still meet its labeled specifications, and the risk that the drug is unsafe or ineffective may be minimal. Thus, the FDA’s advice will be specific to the circumstances, and health care professionals will be best able to balance risks and benefits and make the right decision for their patients.

    If failure to meet cGMP results in the distribution of a defective drug, the company may subsequently recall that product. Removing these drugs from the market will protect the public’s health. While the FDA cannot force a company to recall a drug, companies will usually recall a drug voluntarily or at the FDA’s request. If a company refuses to recall a drug, the FDA can warn the public and could seize the drugs that are on the market.

    Even if the drugs are not defective, the FDA can bring a seizure or injunction case in court to address cGMP violations. When the FDA brings a seizure case, the agency asks the court for an order that allows federal officials to take possession of adulterated drugs and destroy them. This enables the FDA to immediately prevent a company from distributing those drugs to patients. When the FDA brings an injunction case, it asks the court to order a company to stop violating cGMP. Both seizure and injunction cases often lead to court orders that require companies to take many steps to correct cGMP violations, such as hiring outside experts, writing new procedures, and conducting extensive training of their employees. The FDA can also bring criminal cases because of cGMP violations, seeking fines and jail time. Chapter 8 contains more information on this topic.

    Almost every country publishes regulations and guidance documents for the pharmaceutical industry. In the United States they are published in the Federal Register.¹ The FDA’s website (http://www.fda.gov) contains links to the cGMP regulations, guidance documents, and various resources to help drug companies comply with the law. The FDA conducts extensive public outreach through presentations at national and international meetings and conferences to discuss and explain the cGMP requirements, the latest policies, and the agency’s expectations.

    cGMP and the Quality System

    Since the introduction of the ISO 9000 family of quality system standards in 1987, the medical products industry began to look to them for synergies with cGMP regulations. It was clear that regulated businesses can benefit by ensuring the quality of the management system, while cGMP ensures that regulatory requirements are met. Although there was some overlap between the requirements of a QMS and cGMP, they are, in fact, highly complementary.

    Non-US cGMP regulations (EU, Canada, WHO, and so on) have already been harmonized to a QMS, whereas US cGMP never passed through this process. However, in 2006 the FDA made effective the landmark guidance for industry titled Quality Systems Approach to Pharmaceutical CGMP Regulations.² This guidance is intended to help manufacturers implementing effective quality systems and risk management approaches to meet the requirements of the FDA’s cGMP regulations (21 CFR parts 210 and 211). A comparison between US cGMP and QMS elements is presented in the appendix to this book.

    The ICH finalized Q10 Pharmaceutical Quality System in July 2008, and it was officially adopted by the EU in July 2008 and the United States in April 2009.³ This guideline applies to pharmaceutical drug substances and drug products, including biotechnology and biological products, throughout the product life cycle. It is intended to assist pharmaceutical manufacturers by describing a model for an effective QMS for the pharmaceutical industry, referred to as the pharmaceutical quality system. ICH Q10 is discussed in Chapter 4.

    ICH Q10 describes one comprehensive model for an effective pharmaceutical quality system that is based on International Organization for Standardization (ISO) quality concepts, includes applicable cGMP regulations, and complements ICH Q8 Pharmaceutical Development and ICH Q9 Quality Risk Management. ICH Q10 is a model for a pharmaceutical quality system that can be implemented throughout the different stages of a product’s life cycle. Much of the content of ICH Q10 applicable to manufacturing sites is currently specified by national GMP requirements. ICH Q10 is not intended to create any new expectations beyond current regulatory requirements. Consequently, the content of ICH Q10 that is additional to current regional GMP requirements is considered as optional.

    Implementation of ICH Q10 throughout the product life cycle will facilitate innovation and continual improvement and strengthen the link between pharmaceutical development and manufacturing activities. For the purposes of this guidance, the product life cycle includes the following technical activities for new and existing products:

    Pharmaceutical development

    Technology transfer

    Commercial manufacturing

    Product discontinuation

    Following is a discussion of several key QMS concepts as they relate to the manufacture of pharmaceutical products.

    Quality by Design and Product Development

    The concept of quality by design means designing and developing a drug product and associated production processes that will be used during product development to ensure that the product consistently attains a predefined quality at the end of the manufacturing process. Quality by design, along with an effective quality system, provides the framework for the transfer of product knowledge and process understanding from drug development to the commercial manufacturing processes and for post-development changes and optimization. These seminal concepts are main elements within the FDA’s new guidance on process validation.⁴ The cGMP regulations, when viewed in their entirety, integrate the concept of quality by design.

    Quality Risk Management

    Quality risk management is a critical component of an effective quality system framework. It can, for example, help guide the setting of specifications and process parameters for drug manufacturing, assess and mitigate the risk of changing a process or specification, and determine the extent of nonconformance investigations and corrective actions.

    Corrective and Preventive Action

    Corrective and preventive action (CAPA) is a well-known cGMP regulatory concept that focuses on investigating, understanding, and correcting discrepancies and deviations while attempting to avoid their recurrence. Quality system models discuss CAPA as three complementary elements:

    Remedial corrections of an identified problem

    Root cause analysis with corrective action to help understand the root cause of the deviation and avoid recurrence of this cause(s)

    Preventive action to prevent the occurrence of a potential problem

    Change Control

    Change control is another cGMP concept that focuses on managing and controlling changes to prevent unintended consequences. The cGMP regulations provide for change control primarily through the assigned responsibilities of the quality control unit. Certain major manufacturing changes (for example, changes to specifications or a critical product attribute) require regulatory filings and prior regulatory approval. Effective change control activities such as quality planning and control of revisions to specifications, process parameters, and procedures are key components of any quality system.

    The Quality Unit

    Many of the effective quality system concepts correlate closely with the cGMP regulations. Current industry practice generally divides the responsibilities of the quality control unit (QCU), as defined in the cGMP regulations, between quality control (QC) and quality assurance (QA) functions:

    QC usually involves (1) assessing the suitability of incoming components, containers, closures, labeling, in-process materials, and the finished products; (2) evaluating the performance of the manufacturing process to ensure adherence to proper specifications and limits; and (3) determining the acceptability of each batch for release

    QA primarily involves (1) review and approval of all procedures related to production and maintenance, (2) review of associated records, and (3) auditing and performing/evaluating trend analysis

    The QMS term quality unit (QU) is consistent with the cGMP definition in § 210.3(b). The concept of a quality unit is also consistent with modern quality systems in ensuring that the operations associated with all systems are appropriately planned, approved, conducted, and monitored. The cGMP regulations specifically assign the QU the authority to create, monitor, and implement a quality system. Such activities do not substitute the responsibility of other areas (for example, manufacturing personnel) to build quality into the product. The QU should not take on the responsibilities of other units of a manufacturer’s organization, such as the responsibilities handled by manufacturing personnel, engineers, and development scientists.

    Under a quality system, it is necessary that the product and process development units, the manufacturing units, and the QU remain independent.

    Inspection Model

    The FDA’s Drug Manufacturing Inspection Compliance Program,⁷ which contains instructions for FDA personnel for conducting inspections, is a systems-based approach to inspection. For this purpose, the QMS is divided into the following six systems (discussed in detail in Chapter 7):

    Quality system

    Production system

    Packaging and labeling system

    Materials system

    Laboratory controls system

    Facilities and equipment system

    Implementation of a comprehensive quality system model for human and veterinary pharmaceutical products, including biological products, will facilitate compliance with 21 CFR parts 210 and 211. The central goal of a quality system is the consistent production of safe and effective products and ensuring that these activities are sustainable. Quality professionals are aware that good intentions alone will not ensure good products. A robust quality system will promote process consistency by integrating effective knowledge-building mechanisms into daily operational decisions. Specifically, successful quality systems share the following characteristics:

    Science-based approaches

    Decisions based on an understanding of the intended use of a product

    Proper identification and control of areas of potential process weakness

    Responsive deviation and investigation systems that lead to timely remediation

    Sound methods for assessing and reducing risk

    Well-defined processes and products, starting from development and extending throughout the product life cycle

    Systems for careful analysis of product quality

    Supportive management, both philosophically and financially

    Both good manufacturing practice and good business practice require a robust quality system. When fully developed and effectively managed, a quality system will lead to consistent, predictable processes that ensure that pharmaceuticals are safe, effective, and available for the consumer.

    Notes

    Federal Register, https://www.federalregister.gov/.

    US Food and Drug Administration, Guidance for Industry: Quality Systems Approach to Pharmaceutical CGMP Regulations (Rockville, MD: FDA, 2006), http://www.fda.gov/downloads/Drugs/.../Guidances/UCM070337.pdf.

    Food and Drug Administration, Guidance for Industry: Q10 Pharmaceutical Quality System (Washington, DC: FDA, 2009).

    Food and Drug Administration, Guidance for Industry: Process Validation: General Principles and Practices (Washington, DC: FDA, 2011).

    José Rodríguez-Pérez, Quality Risk Management in the FDA-Regulated Industry (Milwaukee, WI: ASQ Quality Press, 2012).

    José Rodríguez-Pérez, CAPA for the FDA-Regulated Industry (Milwaukee, WI: ASQ Quality Press, 2011).

    US Food and Drug Administration, Food and Drug Administration Compliance Program Guidance Manual, Program 7356.002 (Rockville, MD: FDA, 2002), http://www.fda.gov/downloads/ICECI/ComplianceManuals/ComplianceProgramManual/UCM125404.pdf.

    Chapter 2

    US Current Good Manufacturing Practice

    cGMP and the FDA

    In the United States, the production of medical products including drugs is controlled under the federal FD&C Act, which states that a drug product will be deemed to be adulterated unless the methods used in, or the facilities or controls used for, its manufacture, processing, packaging, or holding conform to or are operated or administered in conformity with cGMP. The FDA ensures the quality of drug products by carefully monitoring drug manufacturers’ compliance with its cGMP regulations. The cGMP regulations for drugs contain minimum requirements for the methods, facilities, and controls used in manufacturing, processing, and packing. The regulations make sure that each drug product is safe for use and that it has the ingredients and strength it claims to have.

    The approval process for new drug and generic drug marketing applications includes a review of the manufacturer’s compliance with the cGMP. FDA inspectors determine whether the firm has the necessary facilities, equipment, and skills to manufacture the new drug for which it has applied for approval. Decisions regarding compliance with cGMP regulations are based on inspection of the facilities, sample analyses, and compliance history of the firm. This information is summarized in reports that represent several years of the firm’s history.

    The FDA can issue a warning letter or initiate several other regulatory actions against a company that fails to comply with cGMP regulations. Failure to comply can also lead to a decision by the FDA not to approve an application to market a drug. Chapter 8 expands on this topic.

    The Meaning of Current

    Since the development of the first cGMP regulations for drug products in 1962, the FDA has tried to balance the need for easily understood minimum standards with the need to encourage innovation and the development of improved manufacturing technologies. The FDA gives manufacturers room to determine how to achieve the level of control necessary for cGMP compliance, recognizing that, in some instances, more direction from the agency is necessary to provide a uniform standard to the entire industry, minimize the potential for harm, or achieve some other cGMP objectives. Regulations are periodically reassessed and revised to accommodate advances in technology and other scientific knowledge that further safeguards the drug manufacturing process and the public health.

    In 1996, as part of this reviewing process, the FDA proposed the following significant changes to cGMP regulations:

    Amend certain requirements of the cGMP regulations for finished pharmaceuticals to clarify certain manufacturing, quality control, and documentation requirements and

    Ensure that the regulations more accurately encompass current industry practice (61 FR 20104, May 3, 1996, the 1996 proposed rule)

    Subsequently, as part of the risk-based pharmaceutical cGMP for the twenty-first century initiative, the FDA created the cGMP Harmonization Analysis Working Group (cGMP Working Group) to analyze related cGMP requirements in effect in the United States and internationally, including those related to quality systems. The cGMP Working Group compared parts 210 and 211 (21 CFR parts 210 and 211) with the cGMP of the EU as well as other FDA regulations (for example, the quality system regulation for medical devices, 21 CFR part 820) to identify the differences and consider the value of supplementing or changing the current drug manufacturing regulations. The FDA decided to take an incremental approach in modifying parts 210 and 211 that was based on the cGMP Working Group’s analysis.

    Because of this change in approach, the FDA decided not to finalize the 1996 proposed rule. On December 4, 2007, the FDA published a document withdrawing the 1996 proposed rule (72 FR 68111, the December 2007 proposed rule). On the same date, the agency published a direct final rule (72 FR 68064) and companion proposed rule (72 FR 68113) to clarify and modernize certain provisions of the cGMP

    Enjoying the preview?
    Page 1 of 1