SUMMARY OF THE EMA JOINT REGULATORS/INDUSTRY QBD WORKSHOP LONDON, UK; 28
←
→
Page content transcription
If your browser does not render page correctly, please read the page content below
Downloaded from on January 15, 2021 Summary of the EMA Joint Regulators/Industry QbD workshop (London, UK; 28 −29 January 2014) Graham Cook, Georges France, Øyvind Holte, et al. PDA J Pharm Sci and Tech 2016, 70 163-176 Access the most recent version at doi:10.5731/pdajpst.2015.006171
Downloaded from on January 15, 2021 CONFERENCE REPORT DISCLAIMER: Please note that Conference Reports published in the PDA Journal of Pharma- ceutical Science of Technology are considered as special editorial contributions and are excluded from the Journal’s regular peer review process. This Conference Report is protected by copyright and unauthorized distribution or use is prohibited. CONFERENCE REPORT Summary of the EMA Joint Regulators/Industry QbD workshop (London, UK; 28 –29 January 2014) GRAHAM COOK1,*, GEORGES FRANCE2, ØYVIND HOLTE3, GIAMPIERO LORENTI,4 and DAVID TAINSH5 1 Pfizer UK, Farnham, Surrey, UK; 2Novartis Switzerland, and GSK Switzerland, Prangins (Nyon), Switzerland; 3 Norwegian Medicines Agency, Oslo, Norway; 4Italian Medicines Agency, Rome, Italy; and 5GSK UK, Brentford, Middlesex, UK ©PDA, Inc. 2016 ABSTRACT: This paper summarizes the discussions and insights gained from the key themes that emerged during the Quality by Design (QbD) Workshop held at the European Medicines Agency (EMA) offices in London, UK, on 28 –29 January 2014. Industry and regulators shared practical experiences from six case studies (five approved small molecule products and one phase 3 biotechnological product) based on QbD submissions by five companies (AstraZeneca, GlaxoSmithKline, Novartis, NovoNordisk, and Pfizer). The case studies covered a range of different development, regulatory submission, and post-approval aspects of QbD and were developed through confidential discussions between the company representatives and regulators. Key themes that emerged from the workshop discussions were: 1. presentation of information in submissions (develop- ment story and the presentation of information in marketing authorization applications; risk assessment and criticality); 2. development aspects (design space; use of models; control strategy); and 3. post-approval aspects (lifecycle management; dossier— quality system interactions; handling of deviations). Many aspects of QbD for biotech- nological products are similar to small molecules, but there are some important differences highlighted in this paper. The final section of the paper discusses some proposals for future developments to address the issues that were identified. KEYWORDS: Quality by design (QbD), European Medicines Agency (EMA) Workshop London 2014, Presentation of information, Submissions, Regulatory dossier, Development, Risk assessment, Criticality, Design space, Models, Control strategy, Post-approval, Lifecycle management, Quality system, Deviations. LAY ABSTRACT: This paper summarizes the discussions and insights gained from the key themes that emerged during the Quality by Design (QbD) Workshop held at the European Medicines Agency offices in London, UK, on 28 –29 January 2014. Industry and regulators shared practical experiences from six case studies (five approved small- molecule products and one phase 3 biotechnological product) based on QbD submissions by five companies (AstraZeneca, GlaxoSmithKline, Novartis, NovoNordisk, and Pfizer). The case studies covered a range of different development, regulatory submission, and post-approval aspects of QbD and were developed through confidential discussions between the company representatives and regulators. Key themes that emerged from the workshop discussions were: 1. presentation of information in submissions (develop- ment story and the presentation of information in marketing authorization applications; risk assessment and criticality); 2. development aspects (design space; use of models; control strategy); and 3. post-approval aspects (lifecycle management; dossier— quality system interactions; handling of deviations). Many aspects of QbD for biotech- nological products are similar to small molecules, but there are some important differences highlighted in this paper. The final section of the paper discusses some proposals for future developments to address the issues that were identified. Introduction Workshop, organized by European regulators, in- dustry, and the PDA, at its offices in London. Dur- On 28 –29 January 2014 the European Medicines Agency (EMA) hosted a Quality by Design (QbD) UK; e-mail graham.cook@pfizer.com; telephone ⫹44-7717-878781 * Author for Correspondence: Dr. Graham Cook, doi: 10.5731/pdajpst.2015.006171 12 Greenhill Way, Farnham, Surrey, GU9 8SY, Vol. 70, No. 2, March–April 2016 163
Downloaded from on January 15, 2021 ing the workshop industry and regulators shared The final section of the paper discusses some propos- practical experiences from six case studies based on als for future developments to address the issues that QbD submissions by five companies (AstraZeneca, were identified. GlaxoSmithKline, Novartis, NovoNordisk, and Pfizer). Presentations from the workshop are avail- This paper was developed from summaries of the able on the EMA website QbD page (1), and this different themes prepared by teams of industry work- paper summarizes the discussions and insights shop participants with contributions from the regula- gained from the key themes that emerged during the tory agency participants listed in the Acknowledg- workshop. ments section. The success of the workshop was the result of hard work and enthusiastic contributions by The case studies (five approved small-molecule organizers, case study developers, presenters, and products and one phase 3 biotechnological product) attendees. were chosen to cover a range of different develop- ment, submission, and post-approval aspects of 1. Presentation of Information In Submissions QbD and were developed through confidential dis- cussions between the company representatives and 1.1. The Development Story and Presentation of regulators. Many aspects of QbD for biotechnolog- Information in Marketing Authorization Applications ical products are similar to small molecules and this was reflected in the issues and proposals discussed One of the major issues with science-rich chemistry, during the workshop. However, there are some im- manufacturing and controls (CMC) submissions has portant differences, and those are highlighted in the been the effective communication of the important paper, albeit that to date there has been limited aspects of a development program in a way that ap- experience in Europe with QbD biopharmaceutical propriately differentiates the critical aspects from the submissions. mass of supporting data. Industry and regulators share a common interest in concise, focused submissions The discussion points and recommendations from the that facilitate efficient assessment and support opti- workshop were collated into key themes and grouped mised lifecycle management after approval. Particular into three areas, as follows: aspects of the presentation of information explored during the workshop included: 1. Presentation of information in submissions ● How can the development story in QbD submis- 1.1 Development story and the presentation of in- sions be presented to facilitate assessment? formation in marketing authorization applica- tions ● What details are necessary for review of risk as- sessments, design of experiments (DoEs), process 1.2 Risk assessment and criticality analytical technology (PAT) tools, etc., and how would this vary depending on the use of the infor- 2. Development aspects mation in the finished product design, manufactur- ing process, and control strategy? 2.1 Design space ● Is the common technical document (CTD) format 2.2 Use of models adequate to support presentation and review of enhanced development submissions? Is separation 2.3 Control strategy of registered detail from supportive information in the CTD format clear? 3. Post-approval aspects ● How and where should the control strategy (in- 3.1 Lifecycle management cluding any proposed design space) and its devel- opment be described in the submission? 3.2 Dossier—quality system interactions ● When assessors are considering requests for oper- 3.3 Handling of deviations ational flexibility would it be helpful to describe 164 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 elements of the quality system in the application the degree to which commitments in the manufactur- file to aid understanding of how changes are fore- ing process and control strategy could be thought of as seen to be managed during the lifecycle? non-traditional because it is likely that more under- standing (science and risk/criticality based) would be Recommendations expected in such cases, for example, if a flexible manufacturing process and/or a real-time release test- Information presented in a dossier will vary according ing (RTRt) or similar control strategy is applied, more to the product type and complexity, the nature of the understanding is expected to be presented under de- challenges overcome during development, and the velopment. It was agreed that Pareto plots and knowledge gained—it is not one-size-fits-all. Interna- Ishikawa/fishbone diagrams are useful tools for com- tional Conference on Harmonization (ICH) terminol- municating some risk assessments. If a design space is ogy should be used and non-ICH terminology such as proposed, its development should be clearly described “key”, “major”, or “minor” should be avoided as the in the relevant development sections (e.g., S.2.6/ lack of agreed definition can create confusion. P.2.3), and it should be explicitly defined (e.g., by equation, tables, and/or visual representations) in the Assessment is facilitated by summarizing the control manufacturing process description sections of the dos- strategy that assures delivery of the critical quality sier (e.g., S.2.2/P.3.3). attributes (CQAs) and explicitly stating the extent of flexibility proposed by the applicant. The CTD struc- 1.2. Risk Assessment and Criticality ture is not optimal for provision of this overall per- spective to the assessor, but experience of the work- Communication of risk assessment/management in shop participants suggests that the limitations can be regulatory submissions presents a number of chal- overcome. Sections S.2.6 and P.2 have been used lenges, including: successfully for providing summary information. This control strategy summary would also be useful for the ● Translation of the raw quality risk assessment out- inspector. Tabular formats are recommended to pres- comes into an appropriate summary. ent important elements of the control strategy, linking the quality target product profile (QTPP) to CQAs of ● The difficulty in defining the threshold for “criti- drug product and drug substance and to critical pro- cal” given that it is perceived as a continuum in cess parameters (CPPs) (and material inputs) of the ICH guidance (e.g., ICH Q11 Section 10.2 exam- process. ple 2). The presentation of enhanced or QbD aspects should ● The provision of linkages between risk assessment include sufficient detail or rationale to explain the and the development of the control strategy. derivation of CQAs and CPPs, and the tabular format can link/reference detailed sections of the CTD where ● The difficulties managing post-approval changes further detail can be found. given the differences in the post-approval frame- works between markets (and potentially the ap- The depth and/or extent of information presented on proval of what is a CPP and non-CPP). risk assessments, DoEs, mathematical models, etc., should depend upon their purpose or use in assuring Furthermore, for biotechnological products additional the quality of the product and associated disposition key differences add to the challenges: decisions. For example, if a DoE was used for screen- ing purposes, summary information could be suffi- ● Immunogenicity is a significant concern for bio- cient. By contrast, a DoE used to establish a design logics—and only limited knowledge exists on the space employed as part of the control strategy would relationship between quality attributes and immu- need considerably more detail. This might include the nogenicity. rationale for the DoE approach, tables of inputs (in- cluding batch size, ranges studied, factors kept con- ● Complexities in biotechnological products can stant during the DoE) and results, data interpretation, lead to uncertainties about the definition of CQAs statistical significance of parameters studied, scale- and difficulties establishing linkages of process dependence, and so on. It may be helpful to consider parameters to CQAs. Vol. 70, No. 2, March–April 2016 165
Downloaded from on January 15, 2021 ● CQAs may not be readily analyzed in the final It should therefore be subject to an ongoing process of product and therefore consistency in the manufac- risk assessment during the lifecycle of the product. turing process becomes an important part of the Risk assessments can be used to support variation control strategy and consequently in-process spec- applications and should be available for inspection. ifications and controls are frequently set based on manufacturing capability as well as safety and The use of prior knowledge of the applicant in the efficacy. assessment of risk is acceptable. Risk assessment out- comes that are not aligned with existing scientific ● Even if defined as non-critical, the inclusion of knowledge should be justified. A variety of tabular non-CPPs in process descriptions for biopharma- presentations of risk assessments were presented in the ceuticals can present a burden in lifecycle man- various case studies at the workshop. A matrix pre- agement because changes currently require prior sentation of CQAs versus manufacturing process steps approval before implementation. can be used to summarize the holistic risks and failure modes across a manufacturing process (see Table I) Recommendations and it was also suggested that more detailed tables presenting CQAs versus process parameters (PPs) and Quality risk assessments can help to prioritize devel- material attributes in each of the process steps could opment activities and identify critical process param- be useful. eters and material attributes potentially affecting prod- uct quality, guiding the establishment of the control A good example for presenting a risk assessment strategy. Risk assessment is an iterative process, being summary table is also presented in the training mate- repeated during development as new knowledge is rial of the ICH Q-IWG on the implementation of Q8, gained, so that the extent to which risks have been Q9, Q10. Scoring and thresholds used to classify the mitigated can be assessed. For this reason the se- risks are provided and risks discussed in the comments quence or time points of the risk assessments during column in Table II. the development should be clearly described. The risk assessment tool used (e.g., failure modes and effects The implications of criticality are explored in more analysis, or FMEA) should be stated and, when a detail in subsequent sections: Design Space (2.1), Role quantitative tool is used, scoring and thresholds used of Models (2.2), Control Strategy (2.3). to classify the risks should be explained. Information on the applicant’s experts who performed the risk 2. Development Aspects assessment is not required in the submission and it was agreed that raw data from the risk assessment were not 2.1. Design Space needed for the assessment of the marketing authoriza- tion application. However, if a novel risk assessment Although the design space concept from the ICH Q8, Q9, is used, a definition of the tool and a sample output Q10 guidelines has been incorporated into the European should be provided. regulatory framework including the EU Guideline on Variations categories, the value of the concept is not The differentiation of critical and non-critical param- being fully realized through the product lifecycle. From eters is generally based upon the severity of impact on a global perspective, depending on the regulatory frame- the CQAs of the drug product or drug substance. work in place, the review outcome and lifecycle man- However, for biopharmaceutical products the impact agement of a product submitted with a design space can of process parameters on process performance indica- have substantially different outcomes. tors, such as yield, can also be important to define. In general the identified CQAs are unlikely to change Aspects that were raised for discussion included: during the product lifecycle, although there can be some uncertainty about some potential CQAs for bio- ● Purpose of the design space and role in the control pharmaceuticals. Because the definition of criticality strategy. of a parameter is based upon current knowledge, it could evolve during the development and commercial ● Clarity in the definition of design space in the manufacturing stages of the product lifecycle as pro- dossier and the operational flexibility being re- cess changes are made, or as new knowledge is gained. quested by the applicant, including 166 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 TABLE I Example of Risk Assessment Summary Table from Case Study 1 Showing Highest Failure Modes in Each Category and Quantitative Scores 䡩 The outcome of multivariate experimentation present is seems that design space can be most use- and the description of processes by design space fully used to support the registration of, for example, compared with proven acceptable ranges (PARs); adaptive processes (that is, adjusting CPPs based on upstream attributes) and for application of RTRt based 䡩 Consequences for regulatory changes when non- on soft sensors (e.g., dissolution) where description of CPPs are included in the design space: a poten- the interactions of material attributes and process pa- tial interpretation of the EU Guideline on Vari- rameters are important in defining how the process and ations categories is that all changes to a design product quality is controlled. space are classified as high-risk changes (Type II variations). Can changes to non-CPPs be Depending on how the design space is derived, it may managed differently than CPPs? include both CPPs and non-CPPs, but in some cases a design space description in a dossier may include only ● Impact of scale on design spaces developed using CPPs and material attributes impacting CQAs. Never- small-scale experiments and verification of design theless, a sufficiently detailed description of the man- space at commercial scale. ufacturing process is required in the dossier and this could also include non-CPPs. Operational flexibility Recommendations associated with the design space should be described by the applicant, and it may be beneficial for the Companies should consider the need to develop a applicant to propose how changes associated with the design space and the way it will be applied in the design space would be managed. For example, a table manufacture of the drug product or drug substance. for quality attributes and process parameters defining Enhanced development (QbD) does not require the the design space with the corresponding ranges, to- development of a design space: this is optional. At gether with a table for quality attributes and process Vol. 70, No. 2, March–April 2016 167
Downloaded from on January 15, 2021 TABLE II Example of Risk Assessment Summary Table (FMEA for Purity Control) from ICH Q-IWG Training parameters not included in the design space with their It was confirmed that it is possible to release batches target values or ranges could be used. Target settings based on compliance with the design space, if this has or operating ranges could also be included to indicate been approved by the competent authority. Soft-sensor the initial region where it is intended to operate the models used for RTRt decisions may be supported by commercial process. However, it should be stressed multivariate data analysis (MVDA) and statistical pro- that movement within an approved design space is a cess control (SPC). deliberate, validated adjustment to the process to make it operate optimally within approved limits and hence 2.2. Role of Models does not require a variation application. Process modelling is an integral part of the QbD Design space verification has been discussed in recent framework. Models can be derived to assist in process guidance from the EMA-FDA (U. S. Food and Drug development, process understanding, design space de- Administration) pilot program for parallel assessment termination, on-going process verification, as well as of QbD applications (Q&A published 4 November process control (feedback or feed-forward) thereby 2013). Verification of the whole design space at com- making modelling part of the product lifecycle. The mercial scale prior to commercialization is not neces- increasing use of modelling and inclusion of model- sary. Verification of a more restricted region, for ex- related information in dossiers has highlighted several ample the operating ranges, may be performed areas where common understanding has not been initially. This should be accompanied by a design achieved, including: space verification protocol, which should justify and describe, applying a risk-based approach, how the 1. Operational flexibility associated with the use of applicant will conduct the design space verification models studies. Where the design space has been established using small-scale experiments and models, the impact 2. Terminology and implications related to some em- of scale must be considered by the applicant, and any pirical models— concepts such as out-of-trend scale-independence justified. These justifications can (OOT) and out-of-specification (OOS) be based on first-principles understanding and/or em- pirical correlations. 3. Terminology related to PAT models 168 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 Recommendations that the control strategy is a central output of enhanced development would lead to optimal consideration of Discussions during the workshop confirmed agree- the following key questions by all parties: ment that the level of detail about the model to include in the dossier should be related to the impact of the ● Are CQAs of the product sufficiently assured by model, that is, high-, medium-, and low-risk models as the proposed control strategy? If not, what addi- per the guidance in the ICH Q-IWG Points to Consider tional controls/commitments (or justification) document. For example, if a multivariate statistical should be provided? process control (MSPC) model is used for monitoring only and not for control purposes, then it can be ● Does risk assessment information need to be de- regarded as a low- or medium- impact/risk model. scribed in more detail to justify any enhanced However, if an MSPC model is used as part of a RTRt (reduced) elements of manufacturing process com- strategy then it could be considered a high-impact mitments or controls? model. ● Does the experimentation conducted support the When models are used in the control strategy the applicant’s proposed manufacturing commitments following aspects should be described in order to and controls (e.g., design space, RTRt, flexible provide clarity for assessment of the dossier: processing)? Is potential risk from development of process at small-scale mitigated by the control ● Is a design space model used as a basis for RTRt? strategy applied? ● Is a PAT tool utilized for release, or for in-process ● Does the control strategy assure quality across the monitoring? lifecycle? If not, what additional controls or change management commitments should be con- ● When used for process monitoring, is the model sidered (e.g., design space verification, non-rou- used for continuous improvement, or as an alter- tine tests, etc.)? native approach to validation? Recommendations ● Are calibration samples and validation samples for spectral calibrations clearly defined? Product release against specification alone does not necessarily assure product quality; an appropriate con- ● What model maintenance plans are in place? Be- trol strategy is necessary. The control strategy pro- cause a model is always provisional, its validity posed should provide assurance of all CQAs and should be confirmed by regular review, and up- should come from process understanding guided by dates made as needed. This process should be risk assessment. A control strategy can utilize end- incorporated in the company’s pharmaceutical product tests, input and intermediate material controls, quality system and is subject to inspection. and process and in-process controls from various types of analytical methodologies. Good manufacturing Assumptions and limitations of any models should be practices (GMPs) provide an underlying support to the explained and justified, as appropriate. The type of product-specific elements of the control strategy. As justification for theoretical models (based on first prin- product and process understanding increases it may ciples) and empirical models could be different. When affect the control strategy implemented: statistical indices are used their meaning (and any assumptions) should be explained. ● A product with enhanced understanding can em- ploy a different approach to controls and manufac- 2.3. Control Strategy turing commitments compared to a product for which less understanding exists (where a more A control strategy derived using a systematic science- traditional control strategy, probably including full and risk-based approach is a key output of an en- end-product testing, may be expected). hanced approach to development, linking process un- derstanding, product and process complexity, and the ● Enhanced knowledge can lead to a control strategy applicant’s manufacturing commitments. Agreeing providing enhanced assurance of quality with re- Vol. 70, No. 2, March–April 2016 169
Downloaded from on January 15, 2021 duced end-product testing and increased opera- When considering changes to manufacturing pro- tional flexibility. cesses, the impact on the control strategy should be assessed and the risk to product quality should be ● The effect of scale should be considered for con- evaluated. The introduction of control strategies de- trol strategies developed from small-scale experi- rived from enhanced approaches, such as RTRt, may ments, although a control strategy can be devel- necessitate some changes— usually relatively mi- oped that operates independently of scale or nor—to existing pharmaceutical quality systems. location in the manufacturing operating space. 3. Post-approval Aspects It is important to show how the applicant has control over what the patient needs. Using product and pro- 3.1. Lifecycle Management cess understanding may enable applicants to balance the level of detail provided about the process in rela- One of the major opportunities for industry and regu- tion to the level of detail provided about associated lators is to define how the understanding of product tests. and process can be used to support risk-based deci- sions to manage changes in a product-specific manner ● When a CQA is controlled by end-product testing, during the product lifecycle. An applicant that has the process elements that provide the CQAs should developed enhanced product and process understand- still be described. The description of these process ing and applied it through the control strategy should elements can be at a level of detail that assures be able to achieve different lifecycle management conditions (for example, lower categories of variations regulatory oversight of significant changes bal- for certain changes) than an applicant without such anced with the risk associated to the unit opera- enhanced understanding. There is a perception that tion, and with the importance of the output(s) from enhanced development approaches are not yet result- this unit operation (e.g., consider criticality of ing in benefits for industry and regulators in a reduc- quality attribute(s) affected by that unit operation). tion in the burden of post-approval regulatory In the workshop an example was presented in a changes. Furthermore, it is becoming clear that differ- case study where the description of the type of mill ences in the post-approval frameworks globally make to be used for the active pharmaceutical ingredient it difficult to interpret how to manage changes asso- (API) was sufficient in the context of the proposed ciated with a design space (for example, see the dis- control strategy for particle size distribution, and cussion above on the inclusion of non-CPPs in a additional details of milling conditions were not design space). needed. Moving toward an approach to post-approval change ● When a CQA is controlled in process, it should management that would enhance lifecycle manage- still be discussed in the tabular summary of control ment (e.g., reduce period for implementation of inno- strategy, even though it may not appear on the final vations, reduce the resources associated with the man- product specification. The omission of any CQA agement of post-approval changes) would be a from the specification should be rationalized. common benefit for industry and regulators. This may require an understanding of how the applicant’s qual- Because acceptance criteria are linked to process op- ity management system can support management of erating conditions, a change requested to an accep- quality and management of change and, in conse- tance criterion of an attribute can change the accept- quence, need some consideration of how assessors and able range of manufacturing parameter(s). Process inspectors work together to bring the full value from boundaries should not be overly constrained (for ex- the application and from the quality system, without ample, by requesting ranges to be tightened on the having to include detailed elements of the quality basis of limited batch data showing good process system in the application file. consistency), as this could have a negative impact on process capability. Conversely, broad operating ranges Recommendations not supported by development work, batch analysis data, and/or scientific rationale should not be pro- As noted earlier, the recommendations concerning the posed. presentation of information in dossier are linked with, 170 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 and could support, the optimization of change man- 3.2. Dossier–Quality System Interactions agement through the product lifecycle. The manufacture of a drug substance and drug product A comprehensive, robust control strategy could facil- must be conducted as described in the approved sub- itate management of certain changes if the control(s) mission and according to current GMPs. The relation- are independent of the change (e.g., change of scale, if ship between the submission and the quality system, the controls applied are independent of scale; note that and the corresponding interaction by assessors and at the time of the workshop changes in batch size of inspectors to provide regulatory oversight, is therefore active substances or finished products are subject to of considerable importance. This is even more relevant variation submission). when enhanced and/or non-traditional controls or manufacturing commitments are approved, and this The potential effect of a change could be influenced was exemplified in the discussion points arising from by the control strategy applied. For example, a several of the case studies: change of a manufacturing operating parameter may have a lower risk if it is complemented by additional ● Would closer interactions between assessors and controls that provide an orthogonal view of the inspectors (and R&D and manufacturing) be consistency and reliability of the output post beneficial for enhanced development submis- change. This is analogous to redundancy engineer- sions, and, if so, how can this be achieved in ing approaches (i.e., duplication of components or practice? system elements to increase reliability) and could enable adjustments in the approach to regulatory ● Should certain elements of the pharmaceutical oversight for such changes. quality system (PQS) be summarized in the appli- cation file to provide context for the commitments Management of some changes is accepted as being that are approved, and, if so, what should be in- solely within the company quality system (subject to cluded? regulatory oversight through inspection), while others should not be subject to prior approval but could be 䡩 The ICH Q-IWG Q&A (approved April 2009) “do and tell” with the dossier content being updated on this aspect was noted: through, for example, an annual reporting mechanism. Q3: Is it necessary to describe the PQS in a It was noted that in some regions, including Europe, regulatory submission? certain prospective changes can be managed by utili- A3: No, however, relevant elements of the PQS zation of post-approval change management plans or (such as quality monitoring system, change con- protocols (PACMPs), which are expected to lead to trol, and deviation management) can be refer- faster and more predictable implementation of the enced as part of the control strategy as support- changes post-approval because the strategy and test to ing information. verify the effect of the change on product quality is agreed upon upfront with regulatory authorities. At the ● Despite the IWG Q&A noted above, many com- time of the workshop the experience with PACMPs in panies are concerned that inclusion of any PQS Europe was still relatively limited. information in the registration dossier will lead to the PQS/quality management system information Where products developed by both traditional and being considered as registered detail or regulatory enhanced approaches are manufactured in the same commitments and therefore subject to regulatory facility, the same change management process and post-approval variations. quality management system will be used to assess changes and potential effect on quality. Where there is ● Implementation of ICH Q10 was intended to com- good product and process understanding (e.g., knowl- plement or enhance GMP by achieving product edge gained from an enhanced development ap- realization, establishing and maintaining a state of proach), the risk associated with a certain change may control, and facilitating continuous improvement, be perceived as being lower than an equivalent change and consequently regulatory procedures should where there is less knowledge of the potential effect of facilitate these processes in a streamlined and that change on product quality. efficient way. Vol. 70, No. 2, March–April 2016 171
Downloaded from on January 15, 2021 Recommendations ● The need to present sufficiently detailed manufac- turing process descriptions (i.e., defining process A stronger reliance on the PQS to exercise the re- parameters, their ranges or target values) in a quired control could facilitate certain types of change submission should be balanced with the consequen- management, deviation control, or selection of starting tial need to submit variations for post-approval materials. The possibility of filing “do and tell” vari- changes, which arises from the interpretation that ations was noted as a positive aspect of the European changes to non-CPPs are subject to Type II vari- regulatory system. Examples in some other regulatory ations, requiring prior approval before a permanent systems outside of Europe were also noted. These change to the limits can be made. include the ability to negotiate additional commit- ments (subject to routine inspections) as part of the Recommendations approval process with the US FDA that are subse- Comments from regulators at the workshop suggested quently handled by the annual reporting process and that aspects of the “QP discretion” reflection paper do not require prior approval. In Japan, Pharmaceuti- may be incorporated into the Annex 16 revision to cals and Medical Devices Agency (PMDA) processes formalize the approach to one-off deviations for QPs. allow for changes to certain operating conditions to be There is a need, however, to extend the QP discretion managed within the company quality system at the around minor deviations where they relate specifically discretion and under the oversight of the quality- to parameters classified as non-CPPs in the registra- responsible person. tion document. 3.3. Handling of Deviations It was agreed that to provide assessors with an holistic and sufficiently detailed view of the manufacturing Practical experience with the commercialization of a process it is necessary to provide CPP and non-CPP number of products developed with enhanced ap- details in the process description. A proposal from proaches and utilizing non-traditional control strate- regulators was to identify non-CPPs in the process gies has led to the identification of a number of issues description in the dossier and to specifically state that around the handling of deviations, particularly in re- future post-approval changes to them would be con- lation to non-CPPs presented in the registration dos- ducted according to a Type IA or Type IB process, sier, and qualified person (QP) discretion. providing clarity and agreement at the point of ap- proval of the dossier. ● Requests from regulators to include details such as high and low limits for non-CPPs as part of the 4. Proposals for Future Developments overall process description in the registration dos- sier were reported in the case studies. This brings 4.1. New ICH Quality Topics non-CPPs within the scope of registered details In reviewing the workshop presentations and discus- that must be complied with for successful batch sions, and recommendations from the participants, it is release. As such they are effectively treated simi- apparent that two issues dominated: larly to CPPs, and deviations of non-CPPs against targets or limits creates unintended investigations. ● Presentation of information in submissions—Clar- It would be beneficial to clarify if the QP has the ifying certain aspects of the required level of detail ability to determine whether those materials are and presentation format to facilitate the prepara- acceptable for use. tion and assessment of marketing authorisation applications. ● The EMA QP reflection paper allows for one-off approval of unplanned deviations, however, there ● Lifecycle management—Gaining value from en- is no allowance for multiple deviations of non- hanced knowledge through the commercial phase CPPs. Furthermore, there is often reluctance for of the product lifecycle by facilitating post-ap- QPs to adopt the discretion option as it has not proval changes. been formally included in official guidance (EU GMP Annex 16 was being revised at the time of The output from the workshop informed the contribu- the workshop). tions of the EMA regulators and Efpia industry repre- 172 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 sentatives in the ICH Informal Quality Discussion during the commercial phase of the product life- Group meeting at ICH Minneapolis (June 2014). At cycle. that meeting a 5-year plan for ICH quality topics was developed that includes the following topics: ● Post-approval change management should be fo- cused on managing risk of impact of the change in ● ICH Q12 Technical and Regulatory Consider- a product-specific way taking account of product ations for Pharmaceutical Product Lifecycle understanding/control strategy applied and com- Management pany quality system (as appropriate under ICH Q8, Q11, Q9, and Q10). ● Quality Overall Summary (QOS) ● Consideration of the use of expanded PACMPs The ICH Q12 concept paper includes a description of that allow for management of a particular change three issues to be addressed: across a portfolio of similar products after initial approval of a PACMP. This could be of value for ● Regulatory dossier (level of detail and definition of API starting material change management ap- regulatory commitment) proaches operated by a company with suppliers, for example. ● PQS aspects (harmonized risk-based change man- agement system and knowledge management sys- The elaboration of the QOS topic (together with ICH tem) Q12) could potentially address some of the points identified during the workshop related to the presen- ● Post-approval change management plans and pro- tation of information in marketing authorization appli- tocols cations including: The elaboration of Q12 is aiming to address many of ● Level of detail in the dossier depends on the pur- the points identified during the workshop related to the pose/use of the information (e.g., on DoEs, mod- lifecycle management of post-approval changes, in- els, risk assessments) and should be balanced cluding: against the proposed control strategy and manu- facturing commitments. The QOS could help to set ● A means of differentiation of commitments (e.g., this balance. within process descriptions) based on understand- ing/residual risk (not the current simple definition ● The control strategy is often a key differentiator of criticality). All discussions on criticality are for products developed by enhanced approaches really discussions on post-approval change man- compared with traditional approaches. Providing a agement. summary in the QOS could emphasise this, espe- cially as the CTD structure allows flexibility for ● This differentiation should be explicitly linked to the location of a control strategy summary in Mod- post-approval change management expectations. ule 3. Variations to non-CPPs, if included in a design space or process description, should be managed ● Improved communication between assessors and as a minor change, in line with the low risk posed applicants can often avoid misunderstanding: (e.g., managed internally within a site PQS, or as a Type IA/IB rather than Type II variation). It is 䡩 The clarity of the applicant’s submission is im- agreed that all parameters do not affect a CQA portant—for an assessor to reach the same con- equally: some have more effect than others and clusions as the applicant the submission content therefore the regulatory framework should accom- has to be clear and compelling. modate differences between CPPs vs non-CPPs for post-approval changes. Clarification of the possi- 䡩 The clarity of assessors’ questions is impor- bility for this within the current regulatory frame- tant—the context of the query and why the work or adjustment to the framework would be additional information is being sought can be appropriate and reflect a risk-based approach very valuable to ensure an optimal outcome linked to criticality for management of changes from the review process. Vol. 70, No. 2, March–April 2016 173
Downloaded from on January 15, 2021 䡩 The value of the EMA peer review process ● Use of Models during assessment of centrally authorized prod- ucts is recognized. 䡩 Concept of external validation—parallel testing: 4.2. Elaboration of Q&As and Revisions to the ICH ▪ The term parallel testing is not defined in “Points to Consider” Document ICH, although it is mentioned in the EMA RTRt guideline. Elaboration of new Q&As (or the modification of the ICH IWG Points to Consider Paper or existing ICH ▪ Parallel testing considerations such as the Q-IWG Q&As) could provide helpful clarification of number of batches to be tested during the certain issues identified in the workshop discussions. parallel testing period, prior to RTRt, will Examples of the topics raised at the workshop that depend on the case, for example differences could benefit from Q&As include, for example: between soft sensor models (e.g., predict dis- solution from process data) versus analyzer- ● Design Space based models (e.g., near infrared (NIR) cali- bration). The amount of data available at the 䡩 Regulatory opportunities/consequences for de- time of filing might differ between a legacy sign space in submissions— how is the design product, where a wealth of process data exists, space being used by the company (in the control and a new product with limited full-scale strategy)? experience. 䡩 The level of scrutiny and the data requirements ▪ In general, the parallel testing strategy should be to support submission of the design space and proposed by the applicant but information to be discussion of the underlying model should re- provided in the dossier versus post-approval flect the role of the design space in quality commitments may need further discussion. assurance, the degree of reliance on the design space to ensure product quality, and the role of ▪ Where RTRt is based on soft-sensor models the design space in the broader control strategy. (i.e., prediction based only on process data) In this way design spaces could be categorized MSPC will probably be required in addition to as high, medium, or low impact in a similar parallel testing. manner to the way models are characterised in the ICH “Points to Consider” document. 䡩 Model verification is discussed in the ICH “Points to Consider” document, but further clar- 䡩 It could be helpful to further explore the defi- ification may be required, for example, related nition and regulatory consequences of design to considerations of laboratory versus commer- spaces compared with PARs in relation to the cial scale. science underpinning them, the control strategy, and level of risk, based on criticality assess- 䡩 When using MVDA models to support RTRt the ment, to product quality. Opportunities to create important aspects that need to be included in the a more risk-based lifecycle change categoriza- submission and aspects subject to inspection tion framework would increase the value of could be clarified (e.g., scale issues, ensuring sharing enhanced development knowledge in a the model can accommodate routine process submission and optimize the value of the design variability). space concept. 䡩 Model maintenance aspects should be managed 䡩 The regulatory nature of a detailed verification within the PQS, provided the change is within protocol in the context of other regulatory doc- the scope of the model. For example, following uments, that is, whether this forms part of the the conclusion of parallel testing the model dossier or whether it is a site-based protocol needs to be adapted in a “standard” way, for available for inspection, may need further har- example, change of rank, pre-treatment, or ad- monization. dition of spectra. 174 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 ● Quality System manufacture— knowledge provides the basis for QP discretion. From the example discussed in 䡩 The IWG Q&A on Quality System information workshop, QPs should be allowed discretion for in the dossier could be further clarified, if this is repeated deviations of low-risk parameters against not addressed fully in ICH Q12. the operating limits recorded in the file, based on an assessment of quality. 䡩 Enhancing the clarity of the information about the proposed control strategy to be included in ● If not fully addressed by ICH Q12, establishing the the submission and the information to be avail- inclusion of PQS details in the registration dossier able for inspection is important to facilitate re- would not subject the PQS to EU variation con- view/approval of the submission and post-ap- trols. This could have application to the ICH Q11 proval changes as part of lifecycle management. Q&As currently under development— could the requirements of a quality management system Revisions of the ICH Q8, Q9, Q10 “Points to Con- (QMS) be defined that will enable the marketing sider” document could be considered in a number of authorization holder (MAH) to, for example, as- areas including, for example: sess minor changes or starting materials under the QMS? ● Level of detail in submissions: the “H/M/L im- pact” concept described in Use of Models could be ● Some aspects of the regulatory framework may extended in other areas, that is, the level of detail need to be reconsidered (e.g., variations categori- to be included for DoEs, risk assessments, etc., zation) or clarified to facilitate the success of the depends on the purpose/use in the control strategy implementation of ICH Q8 –11. and assurance of quality. Disclaimer ● Risk and criticality: not all critical parameters are equal in terms of their risk to product quality—a The views expressed in this article are the personal position on the relative criticality of a parameter/ views of the author(s) and may not be understood or material attribute/process input and the relation- quoted as being made on behalf of or reflecting the ship to its understanding/control/residual risk position of the EMA or one of its committees or should be developed. working parties. ● Risk and criticality: the relationship between risk Acknowledgments and process parameter criticality should be aligned with current regulatory thinking (i.e., criticality is The authors would like to thank the presenters, experts based on the effect of a process parameter on from industry, and regulatory agencies that prepared CQAs and risk mitigation does not change criti- the six case studies presented at the workshop (a full cality). list can be found within each case study presentation), the workshop attendees for their participation, and 4.3. Areas for Further Discussion especially the following for preparing reports of the various themes from the workshop from which this Within the EU regulatory framework there are a num- summary paper was prepared: Theodora Kourti ber of aspects that could be explored to resolve some (GSK), Lorenz Liesum (Novartis), Frank Montgomery specific regional aspects discussed at the workshop. (AstraZeneca), Henrik Nielsen (Novo Nordisk), Ron These include: Ogilvie (Pfizer), Preben Østfeldt (Novo Nordisk), Thorbjørn Strøm-Hansen (Novo Nordisk), Gert ● EU GMP Annex 16 and opportunities for QP dis- Thurau (Roche). cretion in relation to the handling of low risk non-CPPs, particularly in assessing quality against The authors would also like to acknowledge the Steer- the operating limits recorded in the file, especially ing Committee members: David Cockburn (EMA), for potential insignificant deviations associated Graham Cook (Pfizer), Lina Ertlé (Roche), Georges with non-CPPs. A key principle is using product/ France (Novartis), Evdokia Korakianiti (EMA), Ric- process understanding to resolve issues during cardo Luigetti (EMA), Sylvie Meillerais (Efpia), Peter Vol. 70, No. 2, March–April 2016 175
Downloaded from on January 15, 2021 Richardson (EMA), Jean-Louis Robert (LNS, Luxem- vid Tainsh (GSK). The following authors declare that bourg), David Tainsh (GSK), and the following for they have no competing interests: Øyvind Holte, organization and support during the workshop: Sylvia Giampiero Lorenti. Becker (PDA), Nigel Hamilton (Sanofi), Simona Ke- ckesova (EMA), Catherine McHugh (IMB, Ireland), Reference Georg Roessling (PDA), Pascal Venneugues (EMA). 1. Joint European Medicines Agency/Parenteral Drug Conflict of Interest Declaration Association Quality-by-Design Workshop, Lon- don, 28 –29 January 2014, http://www.ema.europa. The following authors are employees and/or stock eu/ema/index.jsp?curl⫽pages/news_and_events/ shareholders of the following companies: Graham events/2013/12/event_detail_000808.jsp&mid⫽ Cook (Pfizer); Georges France (Novartis; GSK); Da- WC0b01ac058004d5c3 176 PDA Journal of Pharmaceutical Science and Technology
Downloaded from on January 15, 2021 An Authorized User of the electronic PDA Journal of Pharmaceutical Science and Technology (the PDA Journal) is a PDA Member in good standing. Authorized Users are permitted to do the following: ·Search and view the content of the PDA Journal ·Download a single article for the individual use of an Authorized User ·Assemble and distribute links that point to the PDA Journal ·Print individual articles from the PDA Journal for the individual use of an Authorized User ·Make a reasonable number of photocopies of a printed article for the individual use of an Authorized User or for the use by or distribution to other Authorized Users Authorized Users are not permitted to do the following: ·Except as mentioned above, allow anyone other than an Authorized User to use or access the PDA Journal · Display or otherwise make any information from the PDA Journal available to anyone other than an Authorized User ·Post articles from the PDA Journal on Web sites, either available on the Internet or an Intranet, or in any form of online publications ·Transmit electronically, via e-mail or any other file transfer protocols, any portion of the PDA Journal ·Create a searchable archive of any portion of the PDA Journal ·Use robots or intelligent agents to access, search and/or systematically download any portion of the PDA Journal ·Sell, re-sell, rent, lease, license, sublicense, assign or otherwise transfer the use of the PDA Journal or its content ·Use or copy the PDA Journal for document delivery, fee-for-service use, or bulk reproduction or distribution of materials in any form, or any substantially similar commercial purpose ·Alter, modify, repackage or adapt any portion of the PDA Journal ·Make any edits or derivative works with respect to any portion of the PDA Journal including any text or graphics ·Delete or remove in any form or format, including on a printed article or photocopy, any copyright information or notice contained in the PDA Journal
You can also read