In the experimental (non-clinical) research arena, good laboratory practice or GLP is a quality system of management controls for research laboratories and organizations to ensure the uniformity, consistency, reliability, reproducibility, quality, and integrity of products in development for human or animal health (including pharmaceuticals) through non-clinical safety tests; from physio-chemical properties through acute to chronic toxicity tests.[1][2]

GLP was first introduced in New Zealand and Denmark in 1972, and later in the US in 1978 in response to the Industrial BioTest Labs scandal. It was followed a few years later by the Organization for Economic Co-operation and Development (OECD) Principles of GLP in 1992; the OECD has since helped promulgate GLP to many countries.

GLP applies to non-clinical studies conducted for the assessment of the safety or efficacy of products in development (including pharmaceuticals) for people, animals, and the environment.[1] GLP, a data and operational quality system, is not the same as standards for laboratory safety – appropriate gloves, glasses and clothing to handle lab materials safely. The principles of GLP aim to ensure and promote safety, consistency, high quality, and reliability of chemicals in the process of non-clinical and laboratory testing. GLP is not limited to chemicals and also applies to medical devices, food additives, food packaging, colour additives, animal food additives, other non-pharmaceutical products or ingredients, biological products, and electronic products.

History

The historical events leading to the proposal of the Good Laboratory Practice (GLP) regulations are crucial for understanding why these regulations are important for assuring the quality and integrity of nonclinical safety data. These regulations were developed in response to concerns about the reliability and consistency of safety data used in assessing the potential risks of chemicals and products to humans and the environment. The GLP regulations aim to standardize procedures and practices in nonclinical studies to ensure accurate, reliable, and traceable data. This background helps highlight the significance of adhering to GLP standards in research and regulatory contexts[3].

GLP was first introduced in New Zealand and Denmark in 1972.[4]

The GLP regulations were implemented in the United States in response to cases of fraud involving toxicology labs submitting data to the FDA on behalf of pharmaceutical companies. Industrial BioTest Labs (IBT) was the most notable case where thousands of safety tests for chemical manufacturers were either falsely claimed to have been performed or were of such poor quality that police investigators could not determine the extent of the work completed, despite superficially delivering test results as specified in their contracts with the manufacturers[5]. IBT, a contract laboratory based in Northbrook, Illinois, conducted research for the United States government and various chemical and pharmaceutical companies, both from the U.S. and abroad, and submitted toxicology data to several federal agencies, covering a wide range of products including drugs, insecticides, herbicides, food additives, pesticides, cosmetics, and cleaning products[3].

These issues were made public in the hearings at the US Congress, which led to the FDA's publication of Proposed Regulations on GLP in 1976, with establishment of the Final Rule in June 1979 (21 CFR 58). The Environmental Protection Agency (EPA) had also encountered similar problems in data submitted to it, and issued its own draft GLP regulations in 1979 and 1980, publishing the Final Rules in two separate parts (40 CFR 160 and 40 CFR 792) in 1983[6][7][8].

The OECD

Following Decision C(97),186/Final of the OECD Council, data generated in the testing of chemicals in one OECD Member Country, in accordance with OECD Test Guidelines and the Principles of GLP are accepted in all other OECD Member Countries. OECD: ENV/MC/CHEM(98)17 part two

GLP is a quality system concerned with the organizational process and conditions under which non-clinical health and environmental safety studies are planned, performed, monitored, recorded, archived and reported.[9]

GLP principles include

  1. Organization and Personnel
    • Management-Responsibilities
    • Sponsor-Responsibilities
    • Study Director-Responsibilities
    • Principal Investigator-Responsibilities
    • Study Personnel-Responsibilities
  2. Quality assurance program
    • Quality Assurance Personnel
  3. Facilities
    • Test System Facilities
    • Facilities for Test and Reference Items
  4. Equipment, reagents and materials
  5. Test systems
    • Physical/Chemical
    • Biological
  6. Test and reference items
  7. Standard operating procedures
  8. Performance of study
    • Study Plan
    • Conduct of Study
  9. Reporting of results
  10. Archival – Storage of Records and Reports

OECD Guidelines for the Testing of Chemicals

OECD publishes OECD Guidelines for the Testing of Chemicals, which are guidelines that usually have to be followed for GLP compliance. They are widely required by agencies doing risk assessments of chemicals.

U.S. Food and Drug Administration (21 CFR Part 58)

The FDA requires nonclinical studies on new drugs, food additives, and chemicals to assess their safety and potential effectiveness in humans in compliance with 21 CFR Part 58, Good Laboratory Practice for Nonclinical Studies,[10]. These regulations set the standards for conducting nonclinical laboratory studies that support or are intended to support applications for research or marketing permits for products such as food additives, drugs, medical devices, or biological products. Conducting these studies with rigorous adherence to scientific principles and quality control is crucial, as the decisions based on their outcomes directly affect human health and safety[3]. By adhering to the requirements outlined in 21 CFR Part 58, laboratories conducting nonclinical studies can ensure that the data generated are of high quality, reliable, and suitable for submission to the Agency as part of product approval processes. Compliance with GLP regulations helps to protect the safety and welfare of humans and animals involved in studies and contributes to the overall integrity of scientific research in the development of FDA-regulated products.

The FDA's Good Laboratory Practice (GLP) regulations, detailed in 21 CFR Part 58, were first issued on December 22, 1978, and were enacted into law on June 20, 1979. Proposed amendments were introduced in 1984, leading to the publication of revised GLP regulations on September 4, 1987[3].

Since June 20, 1979, the FDA has received many questions about Good Laboratory Practice (GLP) regulations (21 CFR 58). The responses to these inquiries are stored in the Dockets Management Branch (HFA-305) and shared with the Agency's Bioresearch Monitoring (BIMO) program managers and district offices to ensure consistency. Consequently, the US FDA published the 1981 Questions & Answers - Good Laboratory Practice Regulations document to consolidate and clarify these responses. This Q&A document categorizes responses by specific GLP provisions to make them more useful for both the FDA headquarters and field offices.[11]

Proposed amendments were published in the Federal Register on August 24, 2016, which aimed to require a comprehensive quality system approach known as a GLP Quality System to enhance the current quality system approach for nonclinical laboratory studies[12][13]. This system would be mandatory for safety and toxicity studies that support or are intended to support applications or submissions for products regulated by the FDA[14]. Proposed modifications to the GLP Quality System include additional responsibilities for testing facility management and SOP maintenance, along with expanded definitions applicable to all nonclinical laboratory studies, aiming to enhance roles and functions aligned with the revised testing facility definition and to establish a framework for improving data reliability in regulatory decision-making [12][13].

U.S. Environmental Protection Agency[15]

The EPA's Good Laboratory Practice Standards (GLPS) compliance monitoring program guarantees the accuracy and reliability of test data submitted to the Agency to support pesticide product registration under the Federal Insecticide, Fungicide and Rodenticide Act (FIFRA), section 5 of the Toxic Substances Control Act (TSCA), and in accordance with testing consent agreements and rules issued under section 4 of TSCA. The Agency utilizes data obtained from laboratory inspections and audits to oversee the use of pesticides and industrial chemicals[15].

40 CFR Part 160, Good Laboratory Practice Standards pertains specifically to the Good Laboratory Practice (GLP) standards for pesticide chemicals. It establishes the requirements for conducting studies and generating data used for the registration of pesticides under the Federal Insecticide, Fungicide, and Rodenticide Act (FIFRA). This regulation applies primarily to studies conducted to support the registration or re-registration of pesticide products under FIFRA. It includes studies related to human health and environmental effects of pesticides. It focuses specifically on studies related to pesticide products, including toxicity studies, residue chemistry studies, environmental fate studies, and other types of studies required for pesticide registration. It operates within the context of pesticide regulation under FIFRA, which is specific to the registration and use of pesticides in the United States[7].

40 CFR Part 792, Good Laboratory Practice Standards, covers the broader application of GLP standards for nonclinical laboratory studies conducted for assessing the safety or efficacy of chemical substances, including pesticides, under various regulatory programs overseen by the EPA. This regulation applies to nonclinical laboratory studies conducted for various purposes beyond pesticides, encompassing studies related to chemicals, drugs, food additives, and other substances regulated by the EPA. This part has a broader scope and is applicable to a wider range of substances and regulatory programs. It covers a more diverse range of nonclinical studies, including those related to chemical substances other than pesticides. This could include studies conducted for assessing the safety of industrial chemicals, pharmaceuticals, food additives, and other substances subject to EPA regulation. It operates across various regulatory programs within the EPA, reflecting a broader framework for ensuring the quality and reliability of nonclinical study data used in regulatory decision-making [8].

While both 40 CFR Part 160 and 40 CFR Part 792 address GLP standards for laboratory studies, they differ significantly in terms of scope, applicability, and the specific regulatory context in which they operate. Part 160 is tailored to pesticide registration under FIFRA, whereas Part 792 is a more comprehensive framework applicable to a wider range of nonclinical studies conducted for regulatory purposes across different EPA programs[15].

European Union

Since 1987 the European Council had adopted two basic Directives and a Decision relating to the application of the GLP principles. Directive 2004/10/EC has replaced Directive 87/017/EEC as of 11 March 2004; Directive 2004/9/EC has replaced Directive 88/320/EEC as of 11 March 2004.

This directive lays down the obligation of the Member States to designate the authorities responsible for GLP inspections in their territory. It also comprises requirements for reporting and for the internal market (i.e., mutual acceptance of data).

The 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 followed during laboratory inspections and study audits.

  • 89/569/EEC Council Decision of 28 July 1989 on the acceptance by the European Economic Community of an OECD decision / recommendation on compliance with principles of good laboratory practice.

There are also 'Product Oriented Directives' referring to GLP obligations:

  • REACH Regulation of 18 December 2006 and Directive 2006/121/EC of 18 December 2006
  • Medicinal products; Directive 2001/83/EC on the Community code relating to medicinal products for human use of 6 November 2001 as amended by Commission Directive 2003/63/EC
  • Veterinary Medicinal Products; Directive 2001/82/EC of the European Parliament and of the Council of 6 November 2001 on the Community code relating to veterinary medicinal products
  • Cosmetics; Council Directive 93/35/EEC amending for the 6th time directive 76/768/EEC
  • Regulation (EC) No 1831/2003 of the European Parliament and of the Council of 22 September 2003 on additives for use in animal nutrition
  • Directive 89/107/EEC
  • Novel Foods and novel food ingredients; Regulation (EC) No 258/97 of the European Parliament and of the Council of 27 January 1997 concerning novel foods and novel food ingredients
  • Pesticides; Council Directive 91/414/EEC of 15 July 1991 concerning the placing of plant protection products on the market
  • Biocides; Directive 98/8/EC of the European Parliament and of the Council of 16 February 1998 concerning the placing of biocidal products on the market
  • Detergents; Directive 98/8/EC Regulation (EC) No 648/2004 of the European Parliament and of the Council of 31 March 2004 on detergents
  • EC Ecolabel; Commission Decision 2005/344/EC of 23 March 2005; establishing ecological criteria for the award of the Community eco-label to all-purpose cleaners and cleaners for sanitary facilities

In the meantime the EU has concluded Mutual Acceptance Agreements in the area of GLP with Israel, Japan and Switzerland. By means of the Treaty of the European Economic Area of 13 September 1993, the European Regulations and Directives also apply to Iceland, Liechtenstein and Norway.

Non-OECD member countries

An inspection in non-member economies by OECD inspectors will not guarantee that data generated in compliance with GLP will be accepted in other member countries than the one to which they are submitting data and which has thus sent inspectors to verify the accuracy of their compliance statement.

Klimisch score

The Klimisch score system tries to rank the reliability of toxicity studies for use by risk assessors (regulatory agencies). It was published in 1997, by BASF (a chemical company) authors.[16] Studies performed according to GLP are assigned the top rank of 1 (reliable without restriction) and are preferred by agencies. When no GLP study is available for a particular endpoint, a study with a rank of 2 is usually accepted by an agency. Lower ranks typically require a new study to be performed. Klimisch scoring is very widely used in chemical risk assessments. Critics say it is a self-interested bias on objectivity, that a quality system from the regulated party gives their own GLP-complying studies the top rank.[citation needed][further explanation needed]

Automated systems

In many instances, the optimal recommended "no-argument" means of implementing GLP is to develop an automated approach to both sample preparation and sample measurement. GLP compliance then generally entails including an overarching "chain of custody" sample history and data flow, combined with adequate standard operating procedures for calibration and linearization of measuring tools.[citation needed]

Notes and references

  1. ^ a b "Good laboratory practice (GLP) for safety tests on chemicals". Medicines and Healthcare products Regulatory Agency. 20 January 2017. Archived from the original on 3 September 2014. Retrieved 18 February 2011.
  2. ^ "CFR – Code of Federal Regulations Title 21". www.accessdata.fda.gov. Archived from the original on 4 June 2020. Retrieved 1 June 2020.
  3. ^ a b c d Baldeshwiler, Anne (29 July 2003). "History of FDA good laboratory practices". The Quality Assurance Journal. 7 (3): 157–161. eISSN 1099-1786. ISSN 1087-8378 – via Wiley Online Library.
  4. ^ Kevin Robinson for BioPharm International, 1 Aug 2003. GLPs and the Importance of Standard Operating Procedures Archived 21 March 2005 at the Wayback Machine
  5. ^ Schneider, K (Spring 1983). "Faking it: The case against Industrial Bio-Test Laboratories". Amicus Journal. Natural Resources Defence Council: 14–26. Archived from the original on 14 February 2012. Retrieved 16 April 2011.
  6. ^ Staff, World Health Organization (2009) Handbook: Good Laboratory Practice (GLP) Archived 9 February 2021 at the Wayback Machine
  7. ^ a b "GOOD LABORATORY PRACTICE STANDARDS". www.govinfo.gov. Retrieved 17 April 2024.
  8. ^ a b "GOOD LABORATORY PRACTICE STANDARDS". www.govinfo.gov. Retrieved 17 April 2024.
  9. ^ "OECD Principles of Good Laboratory Practice (as revised in 1997)". OECD Environmental Health and Safety Publications. 1. OECD. 1998. Archived from the original on 20 April 2010. Retrieved 15 October 2006.
  10. ^ "CFR - Code of Federal Regulations Title 21". www.accessdata.fda.gov. Retrieved 12 April 2024.
  11. ^ "1981 Questions & Answers - Good Laboratory Practice Regulations". U.S. Food and Drug Administration (published July 2007). 1981 [June 1981]. Retrieved 12 April 2024.{{cite web}}: CS1 maint: url-status (link)
  12. ^ a b "Good Laboratory Practice for Nonclinical Laboratory Studies". U.S. Food and Drug Administration. 26 March 2018. Retrieved 17 April 2024.
  13. ^ a b "Good Laboratory Practice for Nonclinical Laboratory Studies - A Proposed Rule by the Food and Drug Administration on 08/24/2016". Federal Register - The Daily Journal of the United States Government. 24 August 2016. Retrieved 17 April 2024.
  14. ^ "Regulations.gov". www.regulations.gov. Retrieved 17 April 2024.
  15. ^ a b c US EPA, OECA (30 July 2013). "Good Laboratory Practices Standards Compliance Monitoring Program". www.epa.gov. Retrieved 17 April 2024.
  16. ^ Klimisch, HJ; Andreae, M; Tillmann, U (1997). "A systematic approach for evaluating the quality of experimental toxicological and eco-toxicological data". Regul Toxicol Pharmacol. 25 (1): 1–5. doi:10.1006/rtph.1996.1076. PMID 9056496.

See also

Further reading

External links

Notes

This article is a direct transclusion of the Wikipedia article and therefore may not meet the same editing standards as LIMSwiki.