Who is expected to ensure the highest possible standard of quality are maintained in the conduct of a trial?

Overview

What is GxP?

GxP refers collectively to several different types of “good practice” quality guidelines and regulations, each serving a specific purpose. In pharmaceutical product development, these include but are not limited to:

GxP standards broadly cover what are commonly referred to as the “5 Ps”:

  • Procedures
  • Processes
  • People
  • Premises
  • Products

Rigorously following GxP guidelines safeguards consumer health by preventing poor quality and ineffective or adulterated products. 

What are the consequences of GxP noncompliance?

Sponsors and their partners may face serious consequences, including financial penalties and litigation if they do not follow GxP regulations. Sponsors may receive a warning letter if regulators determine they have significantly disregarded federal regulations and potentially endangered public safety; the sponsor must submit a corrective plan and complete the agreed-upon action steps and confirm with the FDA before continuing development.

In more serious instances, a sponsor may have a product recalled if it poses a risk due to adulteration or misbranding (incorrect labeling) or receive a consent decree if they do not correct regulatory violations and the product presents a definite risk to the public. Companies that receive consent decrees must voluntarily cease operations, remedy the issues, and pass verification audits before resuming operations.

What is the importance of having a Quality Management System (QMS) in place to ensure GxP compliance?

A robust QMS supports the underlying principles of GxP — data integrity, traceability, and accountability — and allows you to share files and reports between groups, all in a secure environment. A well-designed quality system gives you the tools to:

  • Define, publish and revise standard operating procedures (SOPs) for each process or functional area
  • Mitigate risk at every phase by identifying and managing corrective and preventive actions (CAPAs) to reduce errors 
  • Control processes to achieve requisite standards in end products and foster continuous improvement
  • Update procedures in response to regulatory changes or new market opportunities
  • Provide documentation promptly during inspection or regulatory audits
  • Record and archive data to verify tasks have been completed; data should be attributable, legible, contemporaneous, original, and accurate (ALCOA)

Good Clinical Practice (GCP)

What is GCP?

GCP is an international ethical and scientific quality standard for clinical trials to ensure the safety of human subjects involved in research developed by the International Council for Harmonisation, which defines global standards for clinical trials involving humans that governments can adapt into regulations.

Applicable U.S. GCP specifications are FDA 21 CFR 50 (protection of human subjects), 21 CFR 54 (financial disclosure), 21 CFR 56 (Institutional Review Boards), and 21 CFR 312 (trial administration for an investigational new drug application), while the EMA provides direction through both its Clinical Trial Directive (Directive 2001/20/EC) and GCP Directive (Directive 2005/28/EC).

What are the core principles of GCP?

  • The anticipated benefits of a clinical trial should always outweigh the risks
  • Trials should be based on sound scientific evidence, observe ethical standards and comply with detailed protocols
  • The rights, consent, safety, and well-being of human trial participants are of utmost importance
  • All personnel must have the required education, training, and experience.
  • All data are recorded, managed, and stored to enable accurate reporting, verification, and interpretation
  • The investigational drug should be manufactured in compliance with GMP and used according to protocol

Good Laboratory Practice (GLP)

What is GLP?

GLP is a set of rules and criteria for conducting nonclinical toxicology and other safety studies in support of clinical trials and subsequent marketing applications. Toxicology data form the basis for determining a safe first-in-human dose of an investigational drug. GLP ensures that the research performed meets a minimum standard to protect the safety of human subjects.

In the United States, GLP specifications are defined under FDA 21 CFR 58. Comparable European Medicines Agency directives and Organisation for Economic Co-operation and Development principles apply in the European Union.

What are the core principles of GLP?

  • Resources: The R&D organizational structure and responsibilities should be clearly defined with adequate qualified, trained staff; facilities and equipment must be sufficient for the purpose
  • Characterization: For studies designed to evaluate the properties of pharmaceutical compounds, staff must have details about the investigational drug and system used in testing
  • Rules: The study protocol must be documented, and written SOPs are required to provide the technical detail for testing
  • Results: The final report should define how the study was performed, explain scientific data interpretation, and present conclusions  
  • Quality Assurance (QA): QA staff monitoring compliance must be independent of personnel conducting the study 

Good Manufacturing Practice (GMP)

What is GMP?

GMP spells out requirements for the production of investigational drugs and biologics to assure proper identification, quality, purity, and strength for patient safety and efficacy.

The governing U.S. regulations for GMP are FDA 21 CFR 210-211 (drugs) and 21 CFR 600 (biologics). The EMA has promulgated similar directives for the European Union.

What are the core principles of GMP?

  • SOPs for processes and design specifications for equipment and premises are described and documented
  • Employees have appropriate qualifications and training
  • Manufacturing processes are clearly defined and managed. Process changes are reviewed and validated
  • Manufacturing facilities must maintain a clean, controlled environment
  • Manufacturing data are recorded to confirm specified procedures have been followed, and drug products are of the prescribed quality and quantity; deviations are documented and investigated
  • Quality defects are examined, and actions are taken to prevent a recurrence
  • Historical records for tracing each batch of the product are retained
  • Products are packaged and labeled correctly

Best Practices for GxP Compliance

How can you prepare for and ensure GxP compliance?

You can ensure compliance with GxP provisions and maintain quality assurance by establishing a robust QMS that adheres to FDA regulations. Tips for charting a smooth course include:  

  • Hire employees qualified to maintain the QMS
  • Allocate sufficient staff to manage and meet QMS requirements
  • Develop SOPs that detail control of facilities, equipment, laboratory operations, and data recording
  • Conduct annual employee training on GxP standards and SOPs
  • Challenge the QMS with periodic internal audits:
    • For GCP, audit potential clinical sites before starting trials to certify their readiness and prevent delays   
    • During GLP studies, perform phase audits
    • For GMP, a preapproval inspection (PAI) helps assure the FDA that a named production facility is capable of manufacturing a drug and that submitted data are accurate and complete. Before scheduling a formal PAI, it is highly recommended to have a qualified party challenge your QMS from the regulators’ perspective and perform PAI readiness training for all employees

With deep expertise across all relevant GxPs, Regulatory Professionals, A Division of Premier Research, can help clients implement a comprehensive quality management system, providing quality oversight, ensuring regulatory compliance, and preparing for audits in all phases of drug development. To learn more, click here.

1. Federal Food Drug, and Cosmetic Act (FD&C Act) [September 17, 2007]. 21 USC Chapter 9. http://www.fda.gov/opacom/laws/fdcact/fdctoc.htm and Amendments. http://www.fda.gov/opacom/laws/default.htm#amendments.

2. Code of Federal Regulations – Title 21, parts 50 and 56; Title 45, part 46.

3. The Belmont report: ethical principles and guidelines for the protections of human subjects Bethesda (MD): U.S. National Commission for the Protection of Human Subjects of Biomedical and Behavioral Research. 1978. http://www.nih.gov/grants/oprr/humansubjects/guidance/belmont.htm. [PubMed]

4. International Conference on Harmonization. 2007. [September 17]. http://www.ich.org.

5. Murphy SB. The national impact of clinical cooperative group trials for pediatric cancer. Med Pediatr Oncol. 1995;24:279–80. [PubMed] [Google Scholar]

6. McGregor LM, Metzger ML, Sanders R, Santana VM. Pediatric cancers in the new millennium: dramatic progress, new challenges. Oncology. 2007;21(7):809–20. [PubMed] [Google Scholar]

7. http://www.fda.gov/cber/gdlns/clintrialdmc.pdf.

8. Obligations of clinical investigators 21 CFR, part 312.

9. Elements of Informed Consent 21 CFR, 50.25; 45 CFR, 46.116. http://www.fda.gov/oc/ohrt/irbs/informedconsent.html.

10. Trotti A, Colevas AD, Setser A, et al. CTCAE v3.0: development of a comprehensive system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13(3):176–81. [PubMed] [Google Scholar]

11. Shah S, Weitman S, Langevin AM, Bernstein M, Furman W, Pratt C. Phase I therapy trials in children with cancer. J Pediatr Hematol Oncol. 1998;20(5):431–8. [PubMed] [Google Scholar]

12. Smith M, Bernstein M, Bleyer WA, Borsi JD, Ho P, Lewis IJ, et al. Conduct of Phase I trials in children with cancer. J Clin Oncol. 1998; 16:966–78. [PubMed] [Google Scholar]

13. Lee DP, Skolnik JM, Adamson PC. Pediatric phase I trials in oncology : an analysis of study conduct efficiency. J Clin Oncol. 2005;23(33):8431–41. [PubMed] [Google Scholar]

14. Ungerleider RS, Ellenberg S Berg S. Cancer Clinical Trials: Design, Conduct, Analysis, and Reporting. In: Pizzo P, Poplack D, editors. Principles and Practice of Pediatric Oncology. 4th edition Lippincott, Williams and Wilkins; Philadelphia: 2002. p. 385. [Google Scholar]

15. Weitman S, Ochoa S, Sullivan J, Shuster J, Winick N, Pratt C, et al. Pediatric phase II cancer chemotherapy trials: a Pediatric Oncology Group study. J Pediatr Hematol Oncol. 1997;19(3):187–91. [PubMed] [Google Scholar]

16. Simon R. Optimal two-stage designs for phase II clinical trials. Control Clin Trials. 1989;10(1):1–10. [PubMed] [Google Scholar]

17. Chen QR, Vansant G, Oades K, Pickering M, Wei JS, Song YK, et al. Diagnosis of the small round blue cell tumors using multiplex polymerase chain reaction. J Mol Diagn. 2007;9(1):80–8. [PMC free article] [PubMed] [Google Scholar]

18. Lansky SB, List MA, Lansky LL, Ritter-Sterr C, Miller DR. The measurement of performance in childhood cancer patients. Cancer. 1987; 60(7):1651–56. [PubMed] [Google Scholar]

19. Dagher R, Pazdur R. The Phase III Clinical Cancer Trial. In: Andrews P, Teicher B, editors. Anticancer Drug Development Guide: Preclinical Screening, Clinical Trials and Approval. Humana Press; Totowa: 2004. [Google Scholar]

20. Dagher RN, Pazdur R. Clinical Trial Design and Regulatory Issues. In: Abbruzzese JL, Davis D, Herbst R, editors. Antiangiogenic Cancer Therapy. Taylor and Francis; Boca Raton: 2007. [Google Scholar]

21. Kretchmar CS, Kletzel M, Murray K, Thorner P, Joshi V, Marcus R, et al. Response to paclitaxel, topotecan and topotecan-cyclophosphamide in children with untreated disseminated neuroblastoma treated in an upfront phase II investigational window: a Pediatric Oncology Group study. J Clin Oncol. 2004; 22(20):4119–26. [PubMed] [Google Scholar]

22. Pappo AS, Lyden E, Breitfeld P, Donaldson SS, Wiener E, Parham D, et al. Two consecutive phase II window trials of irinotecan alone or in combination with vincristine for the treatment of metastatic rhabdomyosarcoma: the Children's Oncology Group. J Clin Oncol. 2007; 25(4):362–9. [PubMed] [Google Scholar]

23. Walterhouse DO, Lyden ER, Breitfeld PP, Qualman SJ, Wharam MD, Meyer WH. Efficacy of topotecan and cyclophosphamide given in a phase II window trial in children with newly diagnosed metastatic rhabdomyosarcoma: a Children's Oncology Group study. J Clin Oncol. 2004;22(8):1398–1403. [PubMed] [Google Scholar]

24. Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. National Cancer Institute 2003. http://ctep.cancer.gov/forms/CTCAEv3.pdf.

25. Guidance for Industry : Reports on the Status of Postmarketing Study Commitments — Implementation of Section 130 of the Food and Drug Administration Modernization Act of 1997. Feb2006. [September 17, 2007]. http://www.fda.gov/ohrms/dockets/98fr/99n-1852-gdl0002.pdf.

26. Guidance for Industry: Qualifying for Pediatric Exclusivity Under Section 303A of the FD&C Act 1999. Sep1999. [September 17, 2007]. http://www.fda.gov/cder/guidance/2891fnl.htm.

27. Regulations Requiring Manufacturers to Assess the Safety and Effectiveness of New Drugs and Biological Products in Pediatric patients. 63 Federal Register 66631. 1998. [PubMed]

28. Pediatric Exclusivity Provision: Status Report to Congress. U.S. Food and Drug Administration. 2001.

29. Best Pharmaceutical for Children Act. Jan 4, 2002. (Public Law No. 107−109; S-1789, 107th Congress)

30. Guidance for Industry: How to Comply with the Pediatric Research Equity Act. U.S. Food and Drug Administration. Sep2005. [September 17, 2007]. http://www.fda.gov/cder/guidance/6215dft.pdf.

31. Hirschfeld S, Ho PT, Smith M, Pazdur R. Regulatory approvals of pediatric oncology drugs: previous experience and new initiatives. J Clin Oncol. 2003;21(6):1066–73. [PubMed] [Google Scholar]

32. Benjamin DK, Smith PB, Murphy D, Roberts R, Mathis L, Avant D, et al. Peer-reviewed publication of clinical trials completed for pediatric exclusivity. JAMA. 2006;296(10):1266–73. [PMC free article] [PubMed] [Google Scholar]

33. Guidance for Industry: Pediatric Oncology Studies in Response to a Written Request. U.S. Food and Drug Administration. Jun2000. [September 17, 2007]. http://www.fda.gov/cder/guidance/3756dft.htm.

34. European Medicines Agency. Medicines for children. 2007. [September 17, 2007]. http://www.emea.europa.eu/htms/human/paediatrics/introduction.htm.


Page 2

Summary of commonly referenced International Conference on Harmonization (ICH) clinical efficacy guidelines

DocumentSubjectContent
ICH E 2Adverse Event ReportingDefines terms, timeframes for AE reporting and formatting of AE reports
ICH E 6GCP Consolidated GuidelinesDefines responsibilities of sponsors, investigators, consent process monitoring and auditing procedures, and protection of human subjects
ICH E 9Statistical PrinciplesDesign and conduct of trials intended to support or establish efficacy
ICH E 10Choice of Control GroupsProperties and limitations of different kinds of control groups (active control equivalence, non-inferiority, etc.)
ICH E 11Clinical Investigations in Pediatric PopulationPrinciples of clinical investigations in children, including timing of studies and extrapolation of data relative to studies conducted in adults, consent, assent, and interventions
ICH E 14Evaluation of QT/QTc Interval ProlongationTesting the effects of new agents on the QT/QTc interval as well as cardiovascular adverse events