Physical Address
304 North Cardinal St.
Dorchester Center, MA 02124
Good Laboratory Practice (GLP) standards were authored by the United States Food and Drug Administration (FDA) to ensure sound and repeatable nonclinical safety assessment studies. They set the standard by which studies are planned, conducted, monitored, reported, and archived to assure reproducibility, accuracy, and consistency. Similar guidelines were later developed by the United States Environmental Protection Agency (EPA) and internationally by the Organisation for Economic Co-operation and Development (OECD) and regulatory organizations in other countries. The OECD, established in 1961, is an intergovernmental organization with representatives from countries in North America, Europe, and the Pacific Rim as well as the European Commission that promotes policies improving social and economic well-being by coordinating and harmonizing international policies, identification of good practices, discussion of issues of mutual concern, and working collectively to respond to international problems. Over time, the modest differences among GLP standards among various institutions have become increasingly harmonized.
The main goal of GLP in any venue is to assure that animal toxicity studies produce data that are reliable, repeatable, auditable, and globally acceptable. The OECD defines GLP as a “quality system concerned with the organizational process and the conditions under which nonclinical health and environmental safety studies are planned, performed, monitored, recorded, archived, and reported” ( ). GLP attempts to assure integrity, reproducibility, verifiability, and traceability of data. Following GLP principles requires good operational management. Finally, GLP focuses on aspects of study execution including the planning, monitoring, recording, reporting, and archiving that are critical for the reconstruction of the study.
The origin of GLP standards in the United States followed the discovery of scientific misconduct in animal safety testing by pharmaceutical and industrial chemical manufacturers in the 1970s. Before the 1970s, the FDA and EPA received and accepted animal-derived safety data, trusting that the data were generated with integrity. The FDA had gained greater authority of drug safety assessment in 1962 following approval of the Kefauver–Harris Amendments to the Federal Food, Drug, and Cosmetic Act. These amendments followed the thalidomide tragedy in which a sedative widely used outside the United States to treat morning sickness in pregnant women caused birth defects in Europe, Canada, and other countries. The end result of these amendments provided for more safety and efficacy testing as well as better documentation with regular inspection of such records, and afforded the FDA additional time to review new drug applications. In subsequent years, the FDA began requiring drug efficacy data and postapproval reporting.
The United States was not the first to initiate GLP-like standards. New Zealand introduced the Testing Laboratory Registration Act in 1972, defining the “testing laboratory” to encompass staff records, procedures, equipment, and facilities and established a related registration council “to promote the development and maintenance of GLP in testing.” Denmark followed suit that same year. GLP standards in the United States were enacted later that decade following investigation of scientific misconduct.
Between 1975 and 1977 the Senate Judiciary Committee, Subcommittee on Health (chaired by Senator Edward Kennedy), and the FDA investigated multiple incidents of pharmaceutical and contract research organization (CRO) drug testing fraud. Known as the “Kennedy Hearings,” the focus was “Preclinical and Clinical Testing by the Pharmaceutical Industry.” During the hearings, Kennedy stated the following:
Accurate science is the best protection the American people have from an unsafe and ineffective drug supply. Inaccurate science, sloppy science, fraudulent science—these are the greatest threats to the health and safety of the American people. Whether the science is wrong because of clerical error, or because of poor technique, or because of incompetence, or because of criminal negligence is less important than the fact that it is wrong. For if it is wrong, and if, as in this case, the FDA did not—indeed, under the current practice, could not—know it was wrong, then the protective regulatory barrier between a potentially dangerous drug and the patient is removed.
.
G.D. Searle and Company, one of the largest pharmaceutical companies of its time, was a central focus of the investigation. With Searle's full cooperation, the FDA investigated nonclinical safety studies conducted by Searle in house and at a CRO, Hazelton Laboratories, used by Searle. Inspection findings by the FDA investigators included the following:
Excision of tissue masses from rodents during the study and return of those animals to the study with no report of malignant masses to the FDA,
Long delays in reporting “alarming findings” to the FDA,
Lack of histological examination of all collected tissues and gross lesions required by the study protocol,
Differences between initial findings and the pathology raw data submitted in the final report,
Lack of proper personnel training,
Rats listed as dead were recorded later as being alive (sometimes multiple times),
Undocumented study protocol deviations,
Poor accuracy and timeliness of study documentation,
Inconsistent data retention,
No quality control of reported data,
Statistical manipulation to reduce adversity of findings, and
Frank omission of adverse findings from reports.
While criminal charges against employees of Searle were considered, the grand jury decided against indictment. Following the hearings, Searle authored a draft of GLP regulations, submitting it to both the FDA and the Pharmaceutical Research and Manufacturers Association of America, a large portion of which was incorporated into the FDA's draft GLP document. In 1978, the final version of FDA GLP regulations was published in the Code of Federal Regulations (CFR) under Title 21: “Food and Drugs” as Part 58: “Good Laboratory Practice for Nonclinical Laboratory Studies” (21 CFR 58, ), applying to all nonclinical safety studies supporting FDA-regulated products. The FDA Office of Regulatory Affairs (ORA) published “Guidance for Industry GLP Question and Answers” as assistance to research facilities in the interpretation and application of the directives ( ). With standards set for nonclinical safety assessment conduct, the FDA made clear that violating these standards could result in criminal prosecution or disqualification of data and/or the testing facility from conducting further animal testing until violations were remedied. FDA also initiated a more formal inspection and audit program for study examination. The most recent version of FDA GLP was published in 1987 in the Federal Register as “Final Rule—Good Laboratory Practices” ( ). Significant changes were made in the provisions with respect to quality assurance (QA), protocol preparation, test and control article characterization, and retention of specimens and samples based on FDA's experience in implementing the initial 1978 regulations.
Soon after, other agencies released GLP regulations including the EPA and OECD. In 1983, GLP regulations were published by EPA under the Federal Insecticide, Fungicide, and Rodenticide Act (FIFRA; 40 CFR 160, ) and the Toxic Substances Control Act (TSCA; 40 CFR 792, ). In 1981, an international body, the OECD, issued “OECD Principles of Good Laboratory Practice” (revised in 1997, ) and “Guidelines for the Testing of Chemicals” (Test Guidelines, which are periodically reviewed and revised, ). The OECD is not a regulatory agency and does not approve and register new medicines, so its GLP documents are guidelines and not regulations. While the United States is a member of the OECD, the U.S. FDA alone is the sole body for medical product registration in this country; as such, studies conducted for submission to the FDA should be conducted using the FDA GLP standards and not those outlined by the OECD. Fortunately, the increasing harmonization of GLP standards across regulatory agencies around the world over time now permits sponsors and CROs to establish procedures that comply simultaneously with GLP principles and practices of many regulatory bodies ( ).
Following the publication of the FDA GLP standards, the agency formed the Bioresearch Monitoring Program that conducted a pilot inspection program of research facilities to understand their baseline competence level and conformity to GLP standards. The goal of this effort was to prevent fraud, reduce public risk, eliminate the unnecessary use of animals, avoid waste of time and money, avoid acceptance of false results, and ensure confidence in regulatory decisions. “Major failings” noted during the pilot evaluations included lack of QA departments, lack of testing each batch of test article, and failure to maintain standard operating procedures (SOPs) ( ). During the first few investigations, it was found that the industry lacked standards in animal research and testing, quality control measures and their documentation, and recording and reporting of data. Also, important nonclinical safety findings were frequently not reported to the FDA promptly and, occasionally, not reported at all.
After the Kennedy Hearings, scientific misconduct continued to occur, warranting further investigation. In particular, two examples of major scientific fraud occurring in CROs were publicized widely. Biometric Testing Inc. and Industrial Bio-Test Laboratories (IBT) were extensively investigated by the FDA. Biometric Testing Inc. went bankrupt following the guilty pleas of two vice presidents for falsifying reports of animal tests that had not, in fact, been carried out. However, the most sensational case of scientific misconduct was that of IBT, at that time the largest CRO in the world, conducting approximately 35%–40% of all safety studies in the United States. More than 22,000 toxicity studies had been conducted during its existence, mainly supporting the assessment of pesticides and other chemicals, pharmaceuticals, and cosmetics. Violations of sound scientific conduct by IBT included the following:
Poor animal husbandry including constant cage flooding by automatic watering systems resulting in the drowning and frequent escape of test animals,
Faulty record keeping,
Inadequately trained personnel,
“Borrowing” data from control groups of loosely related studies,
Test animals receiving incorrect doses or the wrong test article,
Dead animals listed as alive (similar to Searle),
Chronic studies that reported no findings in control animals despite the geriatric age of the rodents,
Replacement of dead animals mid to late study with naïve animals, with no documentation of this replacement, and
Fabricated data that were never collected.
After the IBT hearings, four senior administrative and technical leaders including the company president were indicted by a U.S. grand jury. Three were found guilty of mail fraud and making a false statement to the government with one sentenced to a year in prison and the other two receiving 6-month sentences. More than 70% of the over 900 studies audited at IBT were invalidated. Sales of marketed products that had been supported by invalidated studies were suspended until new studies were conducted and reviewed.
Following the institution of GLP standards by the U.S. FDA, implementation of GLP standards was broadened by the 1981 OECD decision on Mutual Acceptance of Data (MAD) among member countries ( ) stating that “data generated in the testing of chemicals in an OECD Member country in accordance with OECD Test Guidelines (for chemical testing) and OECD Principles of GLP shall be accepted in other Member countries for purposes of assessment and other uses relating to the protection of man and the environment.” By signing the decision, all OECD member countries agree to conduct all nonclinical safety studies under GLP, advance the international harmonization of GLP and monitoring compliance, and eliminate the need for study repetition to fulfill differing requirements. By instituting international guidance and requirements, international trade in chemicals would not be encumbered by removing the need for duplicative testing. The OECD GLP guidance document provided countries with a framework to implement an internationally recognized standard that still addressed the needs of their own national programs by allowing some minor nation or region-specific variation (i.e., the European Union [EU]), such as archival storage length as an example. Due to the incredible cost and labor associated with chemical testing and the burden of testing in each individual country, OECD sought a way to establish standards that, if followed, would allow acceptance across a number of countries. Barriers to economic progress and world trade were reduced. To meet the conditions of OECD's MAD,
Data must have been produced in compliance with the OECD Principles of GLP, which is assured by the signature of the Study Director,
A functioning regulatory authority must exist in the country where the data are produced, and
The testing facility must be included in the GLP monitoring program of the country's regulatory authority.
Following their acceptance of the OECD GLP Principles, some countries or regions have integrated the Principles into law, as is the case for the EU which adopted the OECD GLP Principles in a European directive.
International organizations and some of their regulations involved in enforcing and modifying GLP guidelines include the following:
Directive 2004/9/EC of the European Parliament and the Council on the inspection and verification of GLP.
This Directive requires that the OECD Revised Guides for Compliance Monitoring Procedures for GLP and the OECD Guidance for the Conduct of Test Facility Inspections and Study Audits must be adhered to during laboratory inspections and study audits.
Directive 2004/10/EC of the European Parliament and the Council on the harmonization of laws, regulations, and administrative provisions relating to the application of the principles of GLP and the verification of their applications for tests on chemical substances.
This directive specifies that Member States must designate the authorities responsible for GLP inspections in their country and outlines requirements for reporting and the internal market (i.e., MAD).
EU: Commission Directive 1999/12/EC of March 8, 1999 adapting to technical progress for the second time the Annex to Council Directive 88/320/EEC on the inspection and verification of GLP.
United Kingdom (UK) Statutory Instrument: “The Good Laboratory Practice Regulations 1997” replaced the existing voluntary UK GLP Compliance program.
European Parliament and the Council of the European Union: 89/569/EEC Council Decision of July 28, 1989 on the acceptance by the European Economic Community of an OECD decision/recommendation on compliance with principles of GLP.
Swiss (GLP/OECD) Working Group on Information Technology (AGIT).
OECD: “OECD Principles of Good Laboratory Practice” [C(97)186(Final)] ( ).
Member nations are comprised of North American, Central American, South American, Asian (including Japan and Korea), and European countries.
Nations currently working with OECD but not as official member countries include Russia, Brazil, China, India, Indonesia, and South Africa.
Chinese Ministry of Science and Technology: Regulation on GLPs.
The initial standard was drafted in 1993, was abolished in 1999, and a new draft was issued following the current U.S. FDA policies when Chinese GLP affairs came under the State Food and Drug Administration (SFDA).
In 2013, the SFDA renamed itself the China Food and Drug Administration (CFDA) to reflect the advancement of the agency to ministerial level. This meant the CFDA would report directly to China's State Council with broader authority to oversee the food, drug, and medical device divisions.
In 2018, in conjunction with China's 2018 government administration overhaul, the CFDA was renamed the National Medical Products Administration (NMPA) and merged into the newly created State Administration for Market Regulation. The NMPA issued regulatory reforms with the goal to improve the drug review process and shorten Investigational New Drug (IND) and New Drug Application (NDA) review timelines, namely by increasing the number of drug reviewers at the Chinese Center for Drug Evaluation. These reforms also aim to encourage and increase novel drug development, accelerate market authorization, and reduce drug lag.
For a complete list of national websites on GLP refer to the OECD website ( ).
Since the February 1, 2020 withdrawal of the UK from the EU, and upon completion of a transition period lasting until the end of 2020, the UK will no longer follow the EU Directives but instead will apply the OECD “MAD” system. The UK as well as many of the EU countries are OECD members already and thus are fully adherent to the MAD system. Therefore, the UK withdrawal from the EU should not have a major impact on the acceptance of GLP data generated in UK facilities by European or other international regulatory receiving authorities.
Established in 1990, the International Conference on Harmonisation (ICH), now referred to as the International Council on Harmonisation, is a collaborative effort to unify and align the drug regulatory authorities and the pharmaceutical industries of Europe, Japan, and the United States. ICH sets forth guidelines on the types and timing of nonclinical safety assessments but does not cover aspects related to the quality or integrity of nonclinical studies as defined by GLPs ( ). They were authored through scientific consensus of regulatory and industry experts, thereby achieving greater harmonization of international pharmaceutical product registration while reducing or obviating duplication of testing carried out during the research and development of new human drugs. The ICH guidelines offer best practices leading to a more economical use of human, animal, and material resources by preventing duplication of human clinical trials, reducing animal usage in safety testing, streamlining regulatory assessment of new drug applications, and reducing time and resources for drug development, ultimately to eliminate unnecessary delays in the availability of new medicines. ICH sets out to remove barriers to global drug development while maintaining the utmost quality, safety, and efficacy to ensure public health. ICH guidelines have been adopted as the regulatory standard in several countries, but are only used as guidance by the FDA to supplement their own existing standard; therefore, familiarity with the content of the ICH guidelines is essential. ICH has published harmonized tripartite guidelines in four areas: quality (chemical and pharmaceutical QA), safety (nonclinical safety assessment), efficacy (clinical research in human patients including Good Clinical Practice [GCP], E6), and multidisciplinary (which includes medical terminology, Medical Dictionary for Regulatory Activities or MedDRA, and Nonclinical Safety Studies, M3, among others).
In 2015, ICH underwent a reformation to extend its global influence beyond the founding regions through formation of the ICH Assembly, an overarching governing body. The Assembly is focused on international pharmaceutical regulatory harmonization that allows pharmaceutical regulatory authorities and other industry organizations to be more actively engaged in ICH's harmonization work. The reforms included the following: increasing global outreach; revamping ICH's governance structure; disseminating more information on ICH processes to a wider number of stakeholders; and establishing ICH as a legal entity to provide for a more stable operating structure.
In December of 2010, the FDA released an Advanced Notice of Proposed Rulemaking to modernize 21 CFR Part 58, the first proposed major revision since 1987 ( ). Incorporating the comments and suggestions of 90 responders, a proposal was opened for comments in early 2017. The proposal addressed nine specific areas of Part 58 including the GLP Quality System, Multisite Studies, Electronic/Computerized Systems, Sponsor Responsibilities, Animal Welfare, Information on QA Inspection Findings, Process-Based Systems Inspections, Test and Control Article Information, and Sample Storage Container Retention. The content of the proposal represents current thoughts and direction of the FDA, but, as a proposal, these concepts are not yet incorporated formally in the enforceable GLP standard.
Highlights of the proposed changes included enhancement of the current quality system approach called the GLP Quality System, conduct and considerations in laboratory oversight (particularly of multisite studies), and harmonization of wording with domestic and international guidelines or regulations, specifically OECD, to increase data integrity and record keeping. The regulations would expand the scope to include tobacco products and veterinary medical devices in addition to those for human use. New and modified definitions, terms, and organizational and personnel roles and responsibilities are detailed including test site, test facility with executive responsibility, attending veterinarian, contributing scientist, and principal investigator for greater clarity. These also included new directives on SOPs for developing, maintaining, and administering SOPs and clarity on management roles, responsibilities, and accountability especially regarding establishing and maintaining the quality system. Additionally, all data generated during a nonclinical study must follow the acronym “ALCOA”: accurate, legible, contemporaneous, original, and attributable. Section 21 CFR 58.61 on equipment design includes computerized systems and other modern equipment used for maintenance, archiving, and retrieval of data since that is currently how most data are stored. To support and expand on the principles of humane animal research, the 3Rs (Replacement, Reduction, and Refinement), the FDA supports the use of nonanimal models when valid alternatives are available. In acknowledgment of the prevalence of multisite studies, the new proposal would revise the Testing Facility definition to “the person responsible for, coordinating, conducting, or completing a nonclinical laboratory study, or any combination thereof” capturing all possible contractual relationships because, in a multisite study, the test facility might not be the facility that conducted the in-life portion of the study as the current definition specifies but rather a contracted or subcontracted person at another location (a satellite entity termed a Test Site). For multisite studies, new communication requirements are added to sponsor responsibilities to establish appropriate lines of communication among all persons conducting any phase of the nonclinical laboratory study as well as documentation by the sponsor. The last goal of the proposed revision is to establish consistency with current domestic and international guidelines, rules, or regulations of GLP ( ). The proposed rule change has generated significant public comment and, as of publication, the proposed changes have not been finalized.
GLP is required only for animal studies conducted to permit safety evaluation of novel products. Categories of products where safety testing is conducted by GLP include food and color additives, human and veterinary drugs, biological products, cosmetics, medical devices, pesticides, electronic products, and cosmetics, as examples. GLPs regulate all nonclinical safety studies that support or are intended to support applications for research or marketing permits for FDA-regulated products, or by similar agencies. GLP standards do not apply to exploratory or efficacy studies, human or animal clinical studies, or chemical analysis. Additionally, repeat-dose pilot toxicity studies and dose escalation studies, while typically conducted in the “spirit” of GLP, are not formally performed under GLP guidelines.
The FDA defines nonclinical studies as any in vivo or in vitro experiment in which test articles are studied prospectively in test systems under laboratory conditions to evaluate their safety. General study types performed under GLP include single-dose toxicity, repeat-dose toxicity, developmental and reproductive toxicity, genotoxicity, carcinogenicity, toxicokinetics, and pharmacodynamics. Phototoxicity, irritation assays, sensitization studies, and abuse potential studies also fall under GLP guidelines. Recall that GLP principles are not directly concerned with study design, which is evaluated by the regulatory authorities (for example, FDA, EPA, OECD), but rather in optimizing the conditions in which the study will be conducted. GLP principles should be applied independently of the site where the study will be performed. For example, if a company outsources a nonclinical study to a CRO or university, those sites must comply with GLP standards to assure acceptance of test results. The main difference between GLP and non-GLP studies is the type and amount of documentation, not in the study design and conduct. GLP studies increase operational costs by up to 30% compared to non-GLP studies. The objective is not only to improve the quality of data but also to maximize the traceability, integrity, accuracy, and reproducibility of data. Similar to GLP studies in animals, clinical trials in humans are regulated by GCP and the production of test articles through Good Manufacturing Practice (GMP).
All GLP standards require a Sponsor-approved study plan or protocol signed by the Study Director, a Quality Assurance Unit (QAU), test article characterization, trained and qualified personnel, procedures for documenting raw data and deviations, a final report, and data archive. Minor differences exist among the standards including the party responsible for compliance (Study Director vs. Test Facility Management), a requirement of a specific statement of GLP compliance in the final report (EPA), laboratory certification and test facility inspection, and archiving process and retention length. In general, institutions that perform animal toxicity studies will design studies so that a single test will address not only the internationally recognized GLP standards but also, if feasible, the minor nation- or region-specific GLP differences that pertain to the markets in which they propose to register their product ( ).
FDA GLP principles are outlined and described in 21 CFR 58. The purpose of this section is to summarize key points of the U.S. FDA regulations, but the OECD and EPA regulations have similar content ( ). Key concepts of the FDA GLP principles include that for each study
Responsibilities are defined for the sponsor management, study management including the study director, and the QAU,
Experimental conduct follows written SOPs,
Testing facilities are adequate to ensure the integrity of a study,
Test and control articles are high quality,
Instruments are calibrated and well maintained,
Personnel are properly trained and educated to execute their duties, and
Raw data are acquired, processed, documented, and archived to ensure the reliability of data.
These principles ensure the uniformity, reproducibility, quality, accuracy, and consistency of study conduct.
This section includes the scope, definitions, applicability to third-party (grant organization and CRO) studies, and test facility inspections. When a sponsor conducting a nonclinical laboratory study that is intended to be submitted to or reviewed by the FDA utilizes the services of a consulting laboratory, contractor, or grantee to perform an analysis or other services, the third party (termed a Test Facility) must be made aware that the study must be conducted in GLP compliance. The responsibility for ensuring compliance with GLP standards rests with the sponsor.
Become a Clinical Tree membership for Full access and enjoy Unlimited articles
If you are a member. Log in here