AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

How does a company demonstrate the implementation of PQS in accordance with ICH?

 regulatory  Comments Off on How does a company demonstrate the implementation of PQS in accordance with ICH?
Oct 132016
 

Image result for Pharmaceutical Quality System

ICH Q10 was published in its final version already in 2008. However, today many companies still have problems to understand how to implement ICH Q10 “Pharmaceutical Quality System” into practice. Quality Assurance and GMP are basic requirements which have been implemented for many years in the pharmaceutical industry (including the API industry). So what is needed to demonstrate that a Pharmaceutical Quality System has been implemented? Please read more about the GMP Questions and Answers.

 

http://www.gmp-compliance.org/enews_05578_How-does-a-company-demonstrate-the-implementation-of-PQS-in-accordance-with-ICH_15515,S-QSB_n.html

ICH Q10 was published in its final version already in 2008. However, today many companies still have problems to understand how to implement ICH Q10 “Pharmaceutical Quality System” in practice. Quality Assurance and GMP are basic requirements which have been implemented for many years in the pharmaceutical industry (including the API industry). So what is needed to demonstrate that a Pharmaceutical Quality System has been implemented?

ICH offers a set of questions and answers which provide more details about the expectations. They were published in 2009 already but are not well-known by the industry. ICH writes: “When implemented, a company will demonstrate the use of an effective PQS through its documentation (e.g., policies, standards), its processes, its training/qualification, its management, its continual improvement efforts, and its performance against pre-defined key performance indicators (see ICH Q10 glossary on performance indicator). A mechanism should be established to demonstrate at a site how the PQS operates across the product lifecycle, in an easily understandable way for management, staff, and regulatory inspectors, e.g., a quality manual, documentation, flowcharts, procedures. Companies can implement a program in which the PQS is routinely audited in-house (i.e., internal audit program) to ensure that the system is functioning at a high level.”

The questions and answers document also states that there is no certification program in place for a Pharmaceutical Quality System. In addition, ICH provides information about how product-related inspections will differ in an ICH Q8, Q9 and Q10 environment. ICH writes: “In the case of product-related inspection (in particular, preauthorization) depending on the complexity of the product and/or process, greater collaboration between inspectors and assessors could be helpful (for example, for the assessment of development data). The inspection would normally occur at the proposed commercial manufacturing site, and there is likely to be greater focus on enhanced process understanding and understanding relationships, e.g., critical quality attributes (CQAs), critical process parameters (CPPs). The inspection might also focus on the application and implementation of quality risk management principles, as supported by the pharmaceutical quality system (PQS).”

In addition to ICH, regulatory authorities also provide further information. The British Authority MHRA, for example, answers the question: Should a company have a procedure to describe how it approaches QRM related to manufacture and GMP? The answer is: “Yes, the procedure should be integrated with the quality system and apply to planned and unplanned risk assessments. It is an expectation of Chapter 1 that companies embody quality risk management. The standard operating procedure (SOP) should define how the management system operates and its general approach to both planned and unplanned risk management. It should include scope, responsibilities, controls, approvals, management systems, applicability, and exclusions.”

The ECA Academy summarised the most relevant questions and answers from regulators like ICH, EMA, FDA etc in a GMP Questions & Answers Guide which allows readers of the document to search for certain GMP questions. A subject index at the beginning of the document lists the most frequent searched terms.

 

//////////PQS,  ICH, Pharmaceutical Quality System

Share

What are the GMP Responsibilities of the Marketing Authorisation Holders?

 regulatory  Comments Off on What are the GMP Responsibilities of the Marketing Authorisation Holders?
Oct 132016
 

 

str1

The European Medicines Agency (EMA) has published a concept paper to summarise the GMP responsibilities of the Marketing Authorisation Holders (MAH).

http://www.gmp-compliance.org/enews_05618_What-are-the-GMP-Responsibilities-of-the-Marketing-Authorisation-Holders_15367,15360,15355,15618,Z-QAMPP_n.html

The GMP/GDP Inspectors Working Group of the European Medicines Agency (EMA) has published a concept paper to summarise the GMP responsibilities of the Marketing Authorisation Holders (MAH). It is not intended to introduce any new responsibilities on MAHs but to document existing requirements in a better way.

The current EU GMP-Guidelines define in several chapters and annexes GMP tasks and responsibilities of the MAH. However, there seems to be a lack of clarity and understanding as to what these responsibilities actually are in their totality, and what they mean for MAHs at a practical level. All these tasks and responsibilities have now been summarised in this concept paper:

  • Chapter 1: responsibility to evaluate the results of the PQR review
  • Chapter 7: responsibility to put contracts in place
  • Chapter 8: responsibilities concerning quality defects, risk-reducing actions and notification of possible disruption in supply
  • Annex 2: responsibility to put contracts in place
  • Annex 12: obligations to approve the design of irradiation cycles, and agreeing the location for retention of irradiation cycle records.
  • Annex 16: requirement to identify the site and QP responsible for certifying each batch (in the case of multiple sites authorised to manufacture / import / certify the same product) and the statement that the “ultimate responsibility for the performance of a medicinal product over its lifetime, its safety, quality and efficacy” lies with the MAH
  • Annex 19: responsibilities for ensuring that reference and retention samples are taken, and stored.

The GMP/GDP Inspectors Working Group thinks that the issues outlined above “are not without important consequences. The way in which MAHs are expected to interact with the manufacturing sites registered in a marketing authorisation is not sufficiently clear, given the diverse ways in which the various MAH responsibilities are set out in the EC Guide to GMP, and differing (often complex) supply chains.”

So what is next?

According to the Concept Paper, “It is recommended that the GMP/GDP Inspectors Working Group (GMP/GDP IWG) should produce a reflection paper intended for Part III of the EU GMP Guide or in another appropriate location (e.g. as proposed by the GMP/GDP IWG). This would capture all of the responsibilities that apply to MAH companies to enable manufacturers to comply with GMP. It would also result in a more complete picture of the regulatory environment with respect to GMP in which the MAH operates.”

The deadline for comments on the concept paper is end of November 2016. Comments should be sent to adm-gmdp@ema.europa.eu.

 

////////GMP Responsibilities,  Marketing Authorisation Holders

Share

How to document a Product Transfer? Example templates!

 regulatory  Comments Off on How to document a Product Transfer? Example templates!
Sep 152016
 

 

str1

All participants of the GMP training course “Product Transfer” will receive a special version of the Guideline Manager CD including documents and templates useable for site change projects.

Click

http://www.gmp-compliance.org/eca_mitt_05359_15221,Z-PEM_n.html

According to the European GMP-Rules, written procedures for tranfser activities and their documentation are required. For example, a Transfer SOP, a transfer plan and a report are now mandatory and will be checked during inspections.

As participant of the GMP education course “Product Transfer” in Berlin, from 25-27 October 2016 you will receive a special version of the Guideline Manager CD with a special section concerning product transfers. This section contains, amongst others, a Transfer SOP and a template for a Transfer Plan. Both documents are in Word format and can immediately be used after adoption to your own situation.

Regulatory Guidance Documents like the WHO guideline on transfer of technology in pharmaceutical manufacturing and the EU/US Variation Guidelines, are also part of the Guideline Manager CD. Due to copyright reasons, this CD is not available for purchase and can only be handed out to participants of the Product Transfer course.

/////////Product Transfer

Share

Analytical Lifecycle: USP <1210> “Statistical Tools”, Analytical Target Profile and Analytical Control Strategy

 regulatory  Comments Off on Analytical Lifecycle: USP <1210> “Statistical Tools”, Analytical Target Profile and Analytical Control Strategy
Sep 152016
 

Image result for statistical tools

Analytical Lifecycle: USP <1210> “Statistical Tools”, Analytical Target Profile and Analytical Control Strategy

The United States Pharmacopeia (USP) is currently undertaking further steps towards a comprehensive analytical lifecycle approach by publishing a draft of a new General Chapter <1210> Statistical Tools for Procedure Validation and two Stimuli Articles regarding Analytical Target Profile and Analytical Control Strategy in Pharmacopeial Forum. Read more about the life cycle concept for analytical procedures.

http://www.gmp-compliance.org/enews_05565_Analytical-Lifecycle–USP–1210–%22Statistical-Tools%22–Analytical-Target-Profile-and-Analytical-Control-Strategy_15438,15608,Z-PDM_n.html

Following the recently announced elaboration of a new general chapter <1220> “The Analytical Procedure Lifecycle” the United States pharmacopeia (USP) is now proceeding in its approach for a comprehensive analytical lifecycle concept. A further step towards this approach is the draft of a new USP General Chapter <1210> Statistical Tools for Procedure Validation which has been published in Pharmacopeial Forum (PF) 42(5) in September 2016. Comment deadline is November 30, 2016.

Additionally, two Stimuli Articles regarding “Analytical Control Strategy” and “Analytical Target Profile: Structure and Application Throughout The Analytical Lifecycle” appeared in the same issue of the PF.

In the draft chapter <1210> Statistical Tools for Procedure Validation, the USP Statistics Expert Committee presents a revision to the proposal of <1210> published in PF 40(5) [Sept.–Oct. 2014]. On the basis of the comments and feedback given by stakeholders, the committee has addressed their concerns about the narrow scope and details on methodology to be used. The chapter is proposed as a companion to general chapter <1225> Validation of Compendial Procedures with the purpose of providing statistical methods that can be used in the validation of analytical procedures. A revision of general chapter <1225>, including a new section on Lifecycle Management of Analytical Procedures, has been published for comment in PF 42(2) in March 2016.

Specifically, the revision clarifies the accuracy and precision calculations while removing specific linearity requirements. Linearity may be inferred from accuracy or other statistical methods as deemed appropriate. The chapter discusses all of the following analytical performance characteristics from a statistical perspective:

  • accuracy,
  • precision,
  • range,
  • detection limit,
  • quantitation limit,
  • and linearity.

Additional related topics that are discussed in the draft include statistical power, two one-sided tests of statistical equivalence, tolerance intervals, and prediction intervals.

Furthermore, up to now, four Stimuli Articles regarding the analytical lifecycle have been published:

and new in PF 42(5)

  • “Analytical Control Strategy”, and
  • “Analytical Target Profile: Structure and Application Throughout The Analytical Lifecycle”.

The Analytical Target Profile (ATP) is the focal point of the lifecycle approach. It is comparable to the Quality Target Product Profile (QTPP) which is defined in ICH Q8. The Stimuli Article emphasizes “that the current approach to development, validation, verification, and transfer of analytical procedures has served the industry well.” The lifecycle approach – comprised of the development (design, stage 1), qualification (stage 2), and monitoring of the performance of analytical procedures (control strategy, stage 3) – is an extension of the current guidance, taking advantage of the learnings from ICH Q8 – quality by design (QbD) concepts. The Article considers in particular the following questions (and provides examples):

  • What is an ATP, and why is it useful?
  • How can the ATP criteria be established?
  • How can an ATP be applied during the three stages of the procedure lifecycle?

According to the article, “an additional advantage of using an ATP is that it can drive the development of a robust control strategy, resulting in better, more consistent performance of an analytical procedure throughout its lifecycle.”

In the Stimuli Article on the Analytical Control Strategy (ACS), the following questions are discussed:

  • What is the ACS?
  • What is the relationship between the ACS and the ATP?
  • What is the quality risk management (QRM) process and how can it be applied to an analytical procedure?
  • How does the ACS apply to the product lifecycle?

Additionally, examples of the following are provided:

  • How to develop an ACS using the QRM process, and
  • How to develop and apply a risk-based replicate strategy to minimize variability.

The USP Expert Panel would appreciate any feedback on the suggested approaches, as well as any alternative approaches for consideration.
Following your registration on the USP Pharmacopeial Forum website you can get to the proposal for general chapter <1210> and the complete stimuli articles. Because of the importance of the new USP chapter <1220>, ECA and USP join forces and organise the first joint event “Lifecycle Approach of Analytical Procedures“, in Prague, Czech Republic, from 8 to 9 November 2016.

 

/////////Analytical Lifecycle,  USP <1210> “Statistical Tools”,  Analytical Target Profile, Analytical Control Strategy

Share

New Q&A Document on the Visual Inspection of Parenterals available

 regulatory  Comments Off on New Q&A Document on the Visual Inspection of Parenterals available
Sep 092016
 

Image result for ECA's Visual Inspection Group

The ECA’s Visual Inspection Group has developed a new document with answers to frequently asked questions. This new Q&A document is now available for download on the Group’s website. Read more about frequently asked questions in the visual inspection.

http://www.gmp-compliance.org/enews_05500_New-Q-A-Document-on-the-Visual-Inspection-of-Parenterals-available_15266,15265,15221,15662,Z-PEM_n.html

The Visual Inspection Group, an Interest Group of the ECA Foundation, has developed a new document with frequently asked questions. The new Q&A document, which was compiled by the Group’s Board, is now available for a free of charge download on the website.

For compiling the document the members of the Group were asked to provide their questions in February. These questions were then evaluated and answered by the Board Members.

The new document is structured as follows:

  • Manual Inspection
  • Automated Inspection
  • Qualification / Validation
  • Test sets
  • Re-Qualification
  • AQL-Tests
  • Defect Categorisation
  • Specific Products
  • Regulatory Affairs
  • Process Control / SPC

Some examples for the questions and the respective answers:

The grey portion of fully automatic control is often checked manually, to return not clearly or fully tested products back to the inspection process. Is it allowed to carry out this testing with the automated inspection machine? From a GMP view, there are no restrictions. It is also important here that at the end a yield calculation and evaluation in the batch record appears. And there are also automated inspection systems that have already integrated the double inspection with multiple cameras.
In highly automated manufacturing lines for LVP flexible containers, the visual inspection process may/cannot comply to the standard visual inspection criteria e.g.: 5 sec inspection time, agitation of the container etc. Is this a compliance problem? The requirements like 5 sec inspection time required by pharmacopoeias are addressing manually performed visual inspection. If the visual inspection is performed automatically, it is the company’s responsibility to ensure that the inspection via camera systems is as effective as a manual visual inspection via a validation (e.g. Knapp Test).

 

Should the AQL be inspected by QC or production AQL manual inspection may be carried out by production staff (to avoid setting up a separate visual inspection team in QC) under a quality oversight or the quality unit. If performed by production operators, the AQL test should not be done by members of the team that was performing the 100 % visual inspection of the batch.

 

The new Q&A document is available for members in the members’ area on the Visual Inspection Group website free of charge. Membership in the Group is also free of charge and merely requires a registration.

The Good Practice Guide “Visual Inspection of medicinal products for parenteral use”, was also revised. The new Version 3.0 will be introduced at the ECA Conference Particles in Parenterals in Barcelona, Spain, from 28-29 September 2016. All delegates of the conference will receive a free copy.

///////// ECA,  Visual Inspection Group, parenteral use, Particles in Parenterals

Share

New Warning Letter of the FDA with the Focus on “Data Integrity”

 regulatory  Comments Off on New Warning Letter of the FDA with the Focus on “Data Integrity”
Sep 092016
 

The FDA has set the focus of its inspections on data integrity for quite some time already. The most recent Warning Letter addressed to a Chinese API manufacturer dated August 2016 clearly concentrates on the topic data integrity. Please find out more about the current FDA Warning Letter in this News.

http://www.gmp-compliance.org/enews_05557_New-Warning-Letter-of-the-FDA-with-the-Focus-on-%22Data-Integrity%22_15488,15844,Z-QCM_n.html

Again, the focus of FDA’s Warning Letter for the Chinese API manufacturer Zhejiang Medicine Co. Ltd. dated 4th August 2016 is on the lack of data integrity. Among other things, records of activities were made not at the time when they have been performed. Moreover, original data have been deleted. A number of alarming findings were discovered in the course of the FDA inspection in June 2015.

The FDA is now expecting concrete measures (“Data Integrity Remediation”) from the company. For this, the FDA expressly recommended to retain qualified, external consultants. Among the measures to be taken:

A – A comprehensive investigation of the extent of incorrect data
1. An extensive plan for the execution of the investigation
2. Interviews of current and former employees to clarify the root cause of incorrect data
3. An assessment of the extent of data integrity deficits.
4. A comprehensive retrospective assessment of the performance of analytical testing.

B – A current risk assessment of the possible effects of the deficiencies identified on the quality of the medicinal products, up to the risk to patients!

C – A management strategy for the implementation of CAPA plans.

All in all, there were great concerns about the authenticity and reliability of the data produced in that company.

To find out more access the complete Warning Letter for Zhejiang Medicine Co. Ltd.

Image result for Zhejiang Medicine Co. Ltd,

//////////Zhejiang Medicine Co. Ltd, Warning Letter,  FDA, Data Integrity

Share

New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach

 Formulation, regulatory  Comments Off on New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach
Sep 022016
 

Image for unlabelled figure

The current work outlines the application of an up-to-date and regulatory-based pharmaceutical quality management method, applied as a new development concept in the process of formulating dry powder inhalation systems (DPIs). According to the Quality by Design (QbD) methodology and Risk Assessment (RA) thinking, a mannitol based co-spray dried formula was produced as a model dosage form with meloxicam as the model active agent.

The concept and the elements of the QbD approach (regarding its systemic, scientific, risk-based, holistic, and proactive nature with defined steps for pharmaceutical development), as well as the experimental drug formulation (including the technological parameters assessed and the methods and processes applied) are described in the current paper.

Findings of the QbD based theoretical prediction and the results of the experimental development are compared and presented. Characteristics of the developed end-product were in correlation with the predictions, and all data were confirmed by the relevant results of the in vitro investigations. These results support the importance of using the QbD approach in new drug formulation, and prove its good usability in the early development process of DPIs. This innovative formulation technology and product appear to have a great potential in pulmonary drug delivery.

Fig. 1

Fig. 1.

Steps and elements of the QbD methodology completed by the authors and applied in the early stage of pharmaceutical development.

“By identifying the critical process parameters, the practical development was more effective, with reduced development time and efforts.”

Edina Pallagi, our QbD evangelist from Hungary shares her team’s experience applying QbD to Dry Powder Inhalation Formulation.

The paper covers:

  • QbD methodology the researchers applied
  • Formulation of dry powder inhalation – API and excipients
  • QTPP, CQA and CPPs  identified for pulmonary use along with target, justification and explanation
  • Characterization test methods
  • Knowledge Space development
  • QbD software used

New aspects of developing a dry powder inhalation formulation applying the quality-by-design approach

  • a Institute of Drug Regulatory Affairs, University of Szeged, Faculty of Pharmacy, Szeged, Hungary
  • b Department of Pharmaceutical Technology, University of Szeged, Faculty of Pharmacy, Szeged, Hungary

///////

Share

FDA published generic user fee for 2017: for ANDA, DMF, and for Facility (API, FDF)

 regulatory  Comments Off on FDA published generic user fee for 2017: for ANDA, DMF, and for Facility (API, FDF)
Aug 032016
 

 

.

 

placeholder+image

http://www.raps.org/Regulatory-Focus/News/2016/07/26/25394/FDA-Lowers-ANDA-Fee-Rates-for-2017/

Generic drugmakers submitting abbreviated new drug applications (ANDAs) and prior approval supplements (PAS) will see their US Food and Drug Administration (FDA) fee rates drop in 2017, though all other rates, including those for drug master files (DMF) and facility fees will increase when compared to 2016.

For FY 2017, the generic drug fee rates are: ANDA ($70,480, down from $76,030 in 2016), PAS ($35,240, down from $38,020 in 2016), DMF ($51,140, up from $42,170 in 2016), domestic active pharmaceutical ingredient (API) facility ($44,234, up from $40,867 in 2016), foreign API facility ($59,234, up from $55,867 in 2016), domestic finished dose formulation (FDF) facility ($258,646, up from $243,905), and foreign FDF facility ($273,646, up from $258,905 in 2016).

The new fees are effective 1 October 2016 and will remain in effect through 30 September 2017.

FDA explained the increases and decreases in fees, noting that for ANDA and PAS fees, the agency is expecting an increase in the number of submissions estimated to be submitted in FY 2017 when compared to 2016. For 2017, the agency estimates that approximately 891 new original ANDAs and 439 PASs will be submitted and incur filing fees.

Fees for DMFs will increase, meanwhile, because of an expected decrease in the number of submissions estimated to be submitted in 2017 (FDA is estimating 379 fee-paying DMFs for 2017), when compared to the estimated submissions from 2016.

And all facility fees will increase in 2017 when compared to the previous year because of a decrease in the number of facilities that self-identified (the total number of FDF facilities identified through self-identification was 675, of which 255 were domestic facilities and 420 foreign facilities; while the total number of API facilities self-identified was 789, of which 101 were domestic facilities and 688 were foreign facilities), FDA said.

How FDA Calculates the Fees

In order to calculate the ANDA fee, FDA estimated the number of full application equivalents (FAEs) that will be submitted in FY 2017, which is done by assuming ANDAs count as one FAE and PASs (supplements) count as one-half of an FAE, since the fee for a PAS is one half of the fee for an ANDA.

The Generic Drug User Fee Act (GDUFA) also requires that 75% of the fee paid for an ANDA or PAS filing be refunded if either application is refused due to issues other than a failure to pay the fees.

And since this is the last year of this iteration of GDUFA (the next version is still in the works), the agency is allowed to further increase the fee revenues and fees established if such an adjustment is necessary to provide for not more than three months of operating reserves for the first three months of FY 2018, though FDA estimates that the GDUFA program will have carryover balances for such activities in excess of three months of such operating reserves, so FDA will not be performing a final year adjustment.

To pay the fees, companies must complete a Generic Drug User Fee Cover Sheet, available at http://www.fda.gov/gdufa and generate a user fee identification (ID) number. Payment must be made in US currency drawn on a US bank by electronic check, check, bank draft, US postal money order or wire transfer.

Federal Register Notice

See more at: http://www.raps.org/Regulatory-Focus/News/2016/07/26/25394/FDA-Lowers-ANDA-Fee-Rates-for-2017/#sthash.FNo99XHR.dpuf

 

 

/////////////FDA,  generic user fee,  2017, ANDA, DMF,  Facility, API, FDF

Share

Written Confirmation expired: Can an API still be imported when produced earlier?

 regulatory  Comments Off on Written Confirmation expired: Can an API still be imported when produced earlier?
Jul 282016
 

 

What needs to be considered if an API is produced in the time period of a valid written confirmation but imported after this confirmation has expired? This is answered in a revised Q&A Document of the EU Commission.

see………http://www.gmp-compliance.org/enews_05432_Written-Confirmation-expired-Can-an-API-still-be-imported-when-produced-earlier_15432,15354,15367,Z-QAMAP_n.html

The EU Commission has updated its Question and Answers Document “Importation of active substances for medicinal products for human use” (now version 7). In this updated version, the question “Can an API batch manufactured during the period of validity of a written confirmation be imported into the EU once the written confirmation is expired?”

In the answer it is referred to Article 46(b)(2)(b) of Directive 2001/83/EC, where it is defined that APIs can only be imported if they are manufactured in accordance with EU GMP or equivalent, and accompanied by a written confirmation from the competent authority of the exporting third country certifying this.

But what if an API is produced in the time period of a valid written confirmation but imported after this confirmation has expired?

In the respective answer the EU Commission states that “it is legitimate to consider that the guarantees of equivalence provided by the written confirmation apply to any API batch in the scope of the written confirmation which was released for sale within the period of validity of the written confirmation, even if not exported in that time period.”

So the answer is ‘yes’, it still can be imported. But it needs to be accompanied by the expired written confirmation together with appropriate documentation which proves “that the whole consignment has been manufactured and released for sale by the quality unit before the expiry date of the written confirmation” and “provides a solid justification of why a valid written confirmation is not available.”

An import without any written confirmation is not possible.

 

///////////API, produced, time period of a valid written confirmation, imported, confirmation has expired, revised Q&A Document of the EU Commission.

Share

Practical Implementation of the Control of Elemental Impurities: EMA’s new Guideline Draft

 regulatory  Comments Off on Practical Implementation of the Control of Elemental Impurities: EMA’s new Guideline Draft
Jul 222016
 

 

One and a half year after its publication, the ICH Q3D guideline still raises many questions. The EMA has recently published a guideline draft aiming at clarifying the practical implementation of ICH Q3D. Read more here about what is expected in a marketing authorisation application or in an application for a CEP with regard to risk assessment and the control of elemental impurities in APIs and medicinal products.

http://www.gmp-compliance.org/enews_05481_Practical-Implementation-of-the-Control-of-Elemental-Impurities-EMA-s-new-Guideline-Draft_15339,15429,15332,S-WKS_n.html

The “ICH Q3D Guideline for Elemental Impurities” was published in December 2014 as Step 4 document and released in August 2015 under No EMA/CHMP/ICH/353369/2013 as EMA’s Scientific Guideline. The guideline came into effect in June 2016 for all medicinal products currently underlying a marketing authorisation procedure (new applications).

In the meantime, it became clear that implementing in practice the requirements of this guideline has been so complex and led to some marketing authorisation procedures being delayed. The ICH has already reacted to the situation and published 7 training modules on its website. Moreover, a concept paper announces a question & answer document.

On 12 July 2016, the draft of an EMA’s guideline entitled “Implementation strategy of ICH Q3D guideline” (EMA/404489/2016) was published. The purpose of the document is to provide support for implementing ICH Q3D in the European context.

The draft comprises three chapters addressing the most important elements in relation with the implementation of the ICH Q3D requirements. The chapter “1. Different approaches to Risk Management” starts describing the two fundamental approaches to the performance of a risk assessment and the justification for a control strategy with regard to elemental impurities:

Drug Product Approach
Here, batches of the finished product are scanned by means of analytical (validated!) procedures to develop a risk-based control strategy. If – with this approach – the omission of a routine testing has to be justified, the authority expects a detailed and valid justification though, and not just analytical data from a few batches.

Component Approach
The guideline draft clearly gives its preference to this approach. The respective contribution of the different components of a medicinal product is considered with respect to the potential total impurity profile and compared to the PDE value from the risk assessment. All potential sources of impurity, for example from production equipment or from excipients of natural (mined) origin have to be considered in this assessment. This particularly applies to outsourced APIs; here, all pieces of information available from Active Substance Master Files (ASMFs) or Certificates of Suitability (CEPs) have to be used. Substances with a Ph.Eur. monograph should always comply with the elemental impurities limits of the corresponding monograph.

The chapter “2. Particulars for Intentionally Added Element(s)” deals with the common practice in many organic syntheses to add elements to increase the specificity of the chemical reaction and the yield. It is particularly critical when the last step of an API synthesis just before the end product uses a metal catalyst. In such a case, the authority expects a convincing evidence that the catalyst is purged to levels consistently below the control threshold (<30% of the PDE) by means of appropriate methods. All details about the API synthesis including the fate of the metals intentionally added have to be consistently described and documented in the marketing authorisation application or in the application for a CEP. If the routine testing of an elemental impurity is needed, the API manufacturer may determine a specification. This information will be required by the medicinal product manufacturer for his overall risk assessment.

The chapter “3. ASMF/CEP: dossier expectations and assessment strategy” explains who has to submit the risk assessment necessary for an ASMF or a CEP and how the dossier will be processed by the assessor of the regulatory authority. Basically, two scenarios are possible:

1. The API manufacturer submits a summary of a risk assessment/management for elemental impurities
Such information flows in the overall risk assessment of the medicinal product manufacturer and is assessed by the quality assessor/ CEP assessor within the marketing authorisation procedure. All data and documents used for the risk assessment should also be available for a GMP inspection.

2. The API manufacturer doesn’t perform any risk assessment/ management.
The regulatory authority basically expects a detailed description of the API synthesis including data on all metal catalysts used. This as well as the analytical routine controls on elemental impurities performed by the API manufacturer will also be assessed by the quality assessor/ CEP assessor. Nevertheless, the assessor won’t make a final conclusion in the ASMF or CEP assessment report with regard to the compliance with ICH Q3D. This will be done within the marketing authorisation procedure for the medicinal product.

The guideline draft can be commented on until 12 August 2016.

///////////ICH Q3D, Control of Elemental Impurities,  EMA, control of elemental impurities in APIs

Share
Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: