Schweitzer Fachinformationen
Wenn es um professionelles Wissen geht, ist Schweitzer Fachinformationen wegweisend. Kunden aus Recht und Beratung sowie Unternehmen, öffentliche Verwaltungen und Bibliotheken erhalten komplette Lösungen zum Beschaffen, Verwalten und Nutzen von digitalen und gedruckten Medien.
Phil Nethercote1 and Joachim Ermer2
1Formerly Analytical Head for the Global Manufacturing and Supply Division of GSK, Luncarty, Perthshire, UK
2Ermer Quality Consulting, Bensheim, Germany
The concept of validation in the pharmaceutical industry was first proposed by two Food and Drug Administration (FDA) officials, Ted Byers and Bud Loftus, in the mid-1970s in order to improve the quality of pharmaceutical products [1]. Validation of processes is now a regulatory requirement and is described in general and specific terms in the FDA's Code of Federal Regulations - CFR21 parts 210 and 211 as well as in the European Medicines Agency (EMA) GMP Guide Annex 15. The 1987 FDA guide to process validation [2] defined validation as "Establishing documented evidence that provides a high degree of assurance that a specific process will consistently produce a product meeting its pre-determined specifications and quality attributes." While the first validation activities were focused on the processes involved in making pharmaceutical products, the concept of validation quickly spread to associated processes including the analytical methods used to test the products.
Regulatory guidance on how analytical methods should be validated has also existed for some time [3]; however, it wasn't until the establishment of the International Conference on the Harmonisation of Technical Requirements for the Registration of Pharmaceuticals for Human Use (ICH) in 1990 that there was a forum for dialogue between regulatory authorities and industry and one of the first topics within the quality section was analytical procedure validation. The ICH was very helpful in harmonizing terms and definitions [4] as well as determining the basic requirements [4]. Of course, due to the nature of the harmonization process, there were some compromises, inconsistencies, and deficiencies, such as the focus on chromatographic methods and chemical actives. Indeed, despite the existence of ICH Q2, a number of regulatory authorities developed their own additional guidance [5, 6].
The recognition that the current Pharmaceutical Industry's manufacturing performance was not as state of the art as other industries [7-9] has resulted in unprecedented efforts over the last 20 years to modernize pharmaceutical development and manufacturing. In August 2002, the FDA announced a significant new initiative to enhance and modernize the regulation of pharmaceutical manufacturing and product quality which resulted in the issue of a report in September 2004 entitled "Pharmaceutical cGMPs for the 21st Century - A Risk Based Approach" [10]. The aims of the initiative included encouraging industry to adopt modern quality management techniques and to implement risk-based approaches that focused both industry and regulatory attention on critical areas. The need to modernize the approach to quality management was also recognized by ICH and resulted in a series of new ICH guidelines being produced. In November 2005, ICH Q8 [11] and Q9 [12] were issued to provide guidance on best practice in pharmaceutical development and risk management. These guidances were followed by ICH Q10 [13] in June 2008, which described the key aspects of a modern pharmaceutical quality system, and by ICH Q11 [14] in May 2012, which gave guidance on the development and manufacture of drug substances. In November 2008, an updated version of ICH Q8 was issued [15], which included an annex that described the concept of Quality by Design (QbD), which was defined as "A systematic approach to development that begins with predefined objectives and emphasizes product and process understanding and process control, based on sound science and quality risk management."
In November 2007, Borman et al. published a paper that recognized that the concepts of QbD, which had been developed with an aim of enhancing the robustness of manufacturing processes, could also have applicability to analytical procedures [16]. The authors noted that the existing guidance on method validation as described by ICH Q2(R1) would need to be substantially rewritten to take account of the QbD risk-based approaches.
FDA had also recognized that existing guidance on manufacturing process validation would need to be revised to better align with modern quality assurance concepts and the report on "Pharmaceutical cGMPs for the 21st Century - A Risk Based Approach" included recommendations that the 1987 industry guideline on process validation be revised to include 21st century concepts, including risk management and adoption of a lifecycle approach. In January 2011, FDA issued a new guidance for industry document entitled "Process Validation: General Principles and Practices" [17]. This guidance aligns process validation activities with a product lifecycle concept and with the ICH Q8, 9, and 10 guidances. The lifecycle concept links product and process development, qualification of the commercial manufacturing process, and maintenance of the process in a state of control during routine commercial production. The FDA guidance revised the definition of process validation to "the collection and evaluation of data, from the process design stage through commercial production, which establishes scientific evidence that a process is capable of consistently delivering quality product" and recognized that process validation involves a series of activities taking place over the lifecycle of the product and process. The guidance describes process validation activities in three stages:
The guideline emphasized that understanding and controlling variation was key to ensuring a process delivered a fit for purpose product. It suggested that manufacturers should:
Control the variation in a manner commensurate with the risk it represents to the process and product and recognized that focusing exclusively on qualification efforts without also understanding the manufacturing process and associated variation may not lead to adequate assurance of quality. It also acknowledged that after establishing and confirming the process, manufacturers must maintain the process in a state of control over the life of the process, even as materials, equipment, production environment, personnel, and manufacturing procedures change.
In 2010, Nethercote et al. [18] suggested that just as process validation can benefit from a product lifecycle approach so can analytical method validation. They also suggested that there were a number of key factors that are important in a QbD/lifecycle approach. These include:
They proposed that method validation be defined as "The collection and evaluation of data and knowledge from the method design stage throughout its lifecycle of use which establishes scientific evidence that a method is capable of consistently delivering quality data," i.e. that, like FDA's definition of process validation it should apply to all activities performed throughout the methods lifecycle of use - not just the qualification step that was traditionally associated the concept of method validation. The only difference being that the output from the method validation activity is the data, whereas from the manufacturing process it is the product. It was also suggested that the three-stage approach defined by FDA could be applied directly to the validation of analytical methods, as illustrated in Figure 1.1. These concepts were further developed in a paper by Nethercote and Ermer in 2012 [19] and by the United States Pharmacopeia (USP) Expert Panel on Validation and Verification [20]. In these papers, the importance of having a well-defined target for the method was emphasized - the concept of having an Analytical Target Profile (ATP) - as well as a recognition that the "Stage 3" activities involved both routine performance monitoring and effective assessment of change.
Figure...
Dateiformat: ePUBKopierschutz: Adobe-DRM (Digital Rights Management)
Systemvoraussetzungen:
Das Dateiformat ePUB ist sehr gut für Romane und Sachbücher geeignet – also für „fließenden” Text ohne komplexes Layout. Bei E-Readern oder Smartphones passt sich der Zeilen- und Seitenumbruch automatisch den kleinen Displays an. Mit Adobe-DRM wird hier ein „harter” Kopierschutz verwendet. Wenn die notwendigen Voraussetzungen nicht vorliegen, können Sie das E-Book leider nicht öffnen. Daher müssen Sie bereits vor dem Download Ihre Lese-Hardware vorbereiten.Bitte beachten Sie: Wir empfehlen Ihnen unbedingt nach Installation der Lese-Software diese mit Ihrer persönlichen Adobe-ID zu autorisieren!
Weitere Informationen finden Sie in unserer E-Book Hilfe.