IMP's Flashcards

1
Q

What are the differences between the various stages of clinical trials?

A

MHRA website guidance for applying for a CTA

(Phase 0 - Microdosing) - small number of human volunteers take 1/100 of the therapeutic dose to obtain Pharmacokinetics information earlier. Primarily to trial safety.

Phase I - (small number of human volunteers). determines the therapeutic range. <typically ‘healthy’ male volunteers> determines dose-effect relationship, side effects and Pharmacokinetics.

Phase II - (several hundred patients). refines dose and length of treatment, identify common side effects. To determine safety and efficacy, bioavailability and proof of concept - Therapeutic Exploratory

Phase III - (several thousand patients), influence the prescribing and patient info. Therapeutic Confirmation, forms basis of MAA. Advantage vs comparator, drug interactions,

Phase IV - (On the marketed product), long term harms and benefits of a medicine and new uses for it. Dose optimisation, drug-drug interactions, additional indications, epidemiology and post-marketing surveillance required by CAs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What changes are there in clinical trials?

A

Clinical Trials Regulation: 536/2014 (Repealing Directive 2001/20/EC) - effective 6 mo after the EC publishes EU CT portal and database (ETA Q3 2019) (APE, ALO!)
- Adverse events reported on a Web based system
- Publically accessible data used to support CTA
- EMA portal for 2 part application and CTA content harmonised (scientific re study and ethics re CT site(s))
- Auxiliary medicinal products replace NIMPs, so don’t cover Non-medicinal agents
- Low intervention CT have less stringent rules re: monitoring and approval timelines - IMP is used per MA and Safety and Efficacy is known.
- Orphan medicinal products (for rare disease) will be fostered

  • Directive 2003/94 (GMP) will be repealed 6 mo after the EC publishes EU CT portal
  • GMP for IMP 2017/1569 Regulation - applies 6 mo after the EC publishes EU CT portal, incorporatessome of the gaps regarding QP certification (536/2014 only requires certification to GMP compliance)
  • Revised annex 13
  • GMP for Human medicinal product 2017/1572 Directive - transpose into national law by 31Mar2018, apply 6 mo after the EC publishes EU CT portal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is an Ethics committee? What do they do and why?

A

Research Ethics Committees (REC) typically in the UK consist of up to 15 members, 1/3rd of whom are lay (broadly, this means their main professional interest is not in a research area, nor are they a registered healthcare professional). They safeguard the rights, safety, dignity and well-being of research participants, independently of research sponsors.

They review applications for research and give an opinion about the proposed participant involvement and whether the research is ethical. RECs are entirely independent of research sponsors (that is, the organisations which are responsible for the management and conduct of the research), funders and investigators. This enables them to put participants at the centre of their review.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is a ‘Clinical trial protocol’?
would you expect to see in this?
What sections are particularly important to the QP?

A

most current definition is in 536/2014:
Protocol = a document that describes the objectives, design, methodology, statistical considerations and organisation of a clinical trial. The term ‘protocol’ encompasses successive versions of the protocol and protocol modifications;

ICH E6(R1) Good Clinical Practice. Protocol contents:
- General Information
- Background information
- Trial Objectives & Purpose
- Trial Design
- Selection and withdrawal of subjects
- Treatment of Subjects
- Assessment of Efficacy
- Assessment of Safety
- Statistics
- Direct Access to Source Data/ Documents
- Quality Control & Quality Assurance
- Ethics
- Data Handling
- Financing & Insurance
- Publication Policy
- Supplements

<underlined>
</underlined>

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is a clinical trial?

A

Eudralex vol 4. Annex 13:
Any investigation in human subjects intended to discover or verify the clinical, pharmacological and/or other pharmacodynamic effects of an investigational product(s) and/or to identify any adverse reactions to an investigational product(s), and/or to study absorption, distribution, metabolism, and excretion of one or more investigational medicinal
product(s) with the object of ascertaining its/their safety and/or efficacy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Supply of packaging materials, how would you sample?

A

Eudralex vol 4. Annex 13:
Instruction to process, package and/or ship a certain number of units of investigational medicinal product(s).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is a ‘low intervention’ clinical trial

A

Eudralex vol 4. Annex 13:
‘Low-intervention clinical trial’ means a clinical trial which fulfils all of the following conditions:
(a) the investigational medicinal products, excluding placebos, are authorised;
(b) according to the protocol of the clinical trial: (i) the investigational medicinal products are used in accordance with the terms of the marketing authorisation; or (ii) the use of the investigational medicinal products is evidence-based and supported by published scientific evidence on the safety and efficacy of those investigational medicinal products in any of the Member States concerned; and
(c) the additional diagnostic or monitoring procedures do not pose more than minimal additional risk or burden
to the safety of the subjects compared to normal clinical practice in any Member State concerned;

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe what is meant by ‘randomisation’ and ‘randomisation codes’

A

Eudralex vol 4. Annex 13:
- Randomisation = The process of assigning trial subjects to treatment or control groups using an element of chance to determine the assignments in order to reduce bias.
- Randomisation Code = A listing in which the treatment assigned to each subject from the randomisation process is identified.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Who is the ‘Sponsor’ in relation to a clincial trial?

A

Eudralex vol 4. Annex 13:
An individual, company, institution or organisation which takes responsibility for the initiation, management and/or financing of a clinical trial.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Who is the ‘Investigator’ in relation to a clincial trial?

A

Eudralex vol 4. Annex 13:
A person responsible for the conduct of the clinical trial at a trial site. If a trial is conducted by a team of individuals at a trial site, the investigator is the responsible leader of the team and may be called the principal investigator.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is an IMP?

A

Eudralex vol 4. Annex 13
IMP = Investigational Medicinal Product
A pharmaceutical form of an active substance or placebo being tested or used as a reference in a clinical trial, including a product with a marketing authorisation when used or assembled (formulated or packaged) in a way different from the authorised form, or when used for an unauthorised indication, or when used to gain further information about the authorised form.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Why is GMP important for IMPs?

A

Eudralex vol 4. Annex 13:
In clinical trials there may be added risk to participating subjects compared to patients treated with marketed products.
The application of GMP is intended to ensure that trial subjects are not placed at risk, and that the results of clinical trials are unaffected by inadequate safety, quality or efficacy arising from unsatisfactory manufacture. Equally, it is intended to ensure that there is consistency between batches of the same IMP used in the same or different clinical trials, and that changes during the development of an IMP are adequately documented and justified.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What are some of the additional risks for IMP/ clinical trials compared to commercial products?

A

Eudralex vol 4. Annex 13:
- Added complexity (lack of fixed routines, variety of clinical trial designs, consequent packaging designs, need, often, for randomisation and blinding, and increased risk of product cross-contamination and mix up.
- May be incomplete knowledge of the potency and toxicity of the product and a lack of full process validation
- Marketed products may be used which have been re-packaged or modified in some way.

These challenges require personnel with a thorough understanding of, and training in, the application of GMP to IMP. Co-operation is required with trial sponsors who undertake the ultimate responsibility for all aspects of the clinical trial including the quality of IMP. The increased complexity in manufacturing operations requires a highly effective quality system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is a NIMP and what will this term change to in new clinical trial regulation?

A

Eudralex vol 4. Annex 13:
NIMP= Non-investigational medicinal product
- Products other than the test product, placebo or comparator which are supplied to subjects participating in a trial. May be used as support or escape medication for preventative, diagnostic or therapeutic reasons and/or needed to ensure that adequate medical care is provided for the subject. They may also be used in accordance with the protocol to induce a physiological response. These products do not fall within the definition of IMP and may be supplied by the sponsor, or the investigator.
The sponsor should ensure that they are in accordance with the notification/request for authorisation to conduct the trial and that they are of appropriate quality for the purposes of the trial taking into account the source of the materials, whether or not they are the subject of a marketing authorisation and whether they have been repackaged. The advice and involvement of a QP is recommended in this task. «< For example: Anticipated emergency situation: A clinical trial where a new biotechnology product is to be given for the first time to humans. The protocol requires availability of appropriate medicinal products needed for the treatment of anaphylactic shock.&raquo_space;»

Redifined in 536/2014 New Clinical Trial Regulation:
Auxiliary medicinal product = a medicinal product used for the needs of a clinical trial as described in the protocol, but not as an IMP.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is ‘reconstition’ in respect of a clinical trial and what authorisations are required for this?

A

Eudralex vol 4. Annex 13:
Both the total and partial manufacture of IMPs, as well as the various processes of dividing up, packaging or presentation, is subject to a manufacturing authorisation. This authorisation, however, shall not be required for reconstitution. For the purpose of this provision, reconstitution shall be understood as a simple process of:
* dissolving or dispersing the IMP for administration of the product to a trial subject,
* or, diluting or mixing the IMP(s) with some other substance(s) used as a vehicle for the purposes of administering it,
(Reconstitution is not mixing several ingredients, including the active substance, together to produce the IMP)

The process of reconstitution has to be undertaken as soon as practicable before administration.
This process has to be defined in the clinical trial application / IMP dossier and clinical trial protocol, or related document, available at the site.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Give examples of clinical trial design

A

ICH Topic E 9: STATISTICAL PRINCIPLES FOR CLINICAL TRIALS
- Parallel Group Design
The most common clinical trial design for confirmatory trials. Subjects are randomised to one of two or more arms, each arm being allocated a different treatment. These treatments will include the investigational product at one or more doses, and one or more control treatments, such as placebo and/or an active comparator.
- Crossover Design
In the crossover design, each subject is randomised to a sequence of two or more treatments, and hence acts as his own control for treatment comparisons. In the simplest 2×2 crossover design each subject receives each of two treatments in randomised order in two successive treatment periods, often separated by a washout period.
- Factorial Designs
In a factorial design two or more treatments are evaluated simultaneously through the use of varying combinations of the treatments. The simplest example is the 2×2 factorial design in which subjects are randomly allocated to one of the four possible combinations of two treatments, A and B say. These are: A alone; B alone; both A and B; neither A nor B. In many cases this design is used for the specific purpose of examining the interaction of A and B.
- Multicentre Trials
Multicentre trials are carried out for two main reasons. Firstly, a multicentre trial is an accepted way of evaluating a new medication more efficiently; under some circumstances, it may present the only practical means of accruing sufficient subjects to satisfy the trial objective within a reasonable time-frame. Multicentre trials of this nature may, in principle, be carried out at any stage of clinical development. They may have several centres with a large number of subjects per centre or, in the case of a rare disease, they may have a large number of centres with very few subjects per centre. Secondly, a trial may be designed as a multicentre (and multi-investigator) trial primarily to provide a better basis for the subsequent generalisation of its findings.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is a CTA and what does it contain?

A

In the UK, a Clinical Trial Authorisation (CTA) from Medicine and Healthcare Products Regulatory Agency (MHRA) is required for a Clinical Trial of an Investigational Medicinal Product (CTIMP). For international trials in Europe, an application to the competent authority in each member state is required.
Contents:
- Section A: Trial Identification*
- Section B: Identification of the sponsor responsible for the request*
- Section C: Applicant Identification
- Section D: Information on each IMP *
- Section E: General Information on the trial*
- Section F: Population of Trial Subjects
- Section G: Proposed Trial Sites/Investigators in the Member State*
- Section H: Competent Authority/Ethics Committee*
- Section I: Signature
- Section J: Check List

  • in IMPD
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is an IMPD and what does it contain?

A

Regulation 536/2014: IMPD= Investigational Medicinal Product Dossier

  • The IMPD shall give information on the quality of any IMP, the manufacture and control of the IMP, and data from non-clinical studies and from its clinical use.
  • The IMPD should be provided in a clearly structured format following the CTD format of Module 3 and include the most up-to-date available information relevant to the clinical trial at time of submission of the clinical trial application.
    Sections are presented for:
  • Drug Substance: Module S (S.1 – General Information, S.2 – Manufacture, S.3 – Characterisation, S.4 – Control of the Drug Substance, S.5 – Reference Standards or materials, S.6 – Container Closure System, S.7 – Stability,
  • Investigational Medicinal Product: Module P (P.1 – Description and Composition, P.2 – Pharmaceutical Development, P.3 – Manufacture, .P.4 – Control of Excipients, P.5 – Control of IMP, P.6 – Reference standards, P.7 – Container closure system, P.8 – Stability
  • Appendices: Module A (<not applicable for IMPD?? A.1 – Facilities and Equipment>, A.2 – Adventitious Agents Safety Evaluation, A.3 – Novel Excipients, A.3 – Novel Excipients,
  • Placebo and equivalent for modified comparator/ reference product: Module P (P.1-8)
  • Placebo: Info as per IMP where relevant. Also cover; masking differences in terms of taste, smell and look, test to differentiate between test and placebo products, shelf life for duration of trial. Stability studies if reason to expect placebo will undergo degradation or changes to key characteristics.
  • Comparator/ Reference Product: Info as per relevant requirements from IMP. Also cover; any additional substances added to authorised product and reference relevant pharmacopoeia or in-house monograph, modifications influencing quality of product (function, safety and efficacy)- demonstrate as comparable. Solid oral dosage forms -comparative dissolution profile, All manufacturing sites, proposed batch formula and modification steps including IPCs, specification and methods; description, ID of API and any other control parameters. If significant modification, consider impact to impurities etc and ensure suitable control. Shelf life for duration of trial. Stability studies if significant modification to comparator.
  • Regional Information: Module R.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is ‘Blinding’

A

Eudralex vol 4. Annex 13:
A procedure in which one or more parties to the trial are kept unaware of the treatment assignment(s). Single-blinding usually refers to the subject(s) being unaware, and doubleblinding usually refers to the subject(s), investigator(s), monitor, and, in some cases, data analyst(s) being unaware of the treatment assignment(s). In relation to an investigational medicinal product, blinding shall mean the deliberate disguising of the identity of the product in accordance with the instructions of the sponsor. Unblinding shall mean the disclosure of the identity of blinded products.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is GCP and what is the QP specifically concerned with in GCP?

A

Regulation 536/2014: GCP = Good Clinical Practice

  • A set of detailed ethical and scientific quality requirements for designing, conducting, performing, monitoring, auditing, recording, analysing and reporting clinical trials ensuring that the rights, safety and well-being of subjects are protected, and that the data generated in the clinical trial are reliable and robust.
  • Principles that have their origin in the World Medical Association’s Declaration of Helsinki.
    (When designing, conducting, recording and reporting clinical trials, detailed questions may arise as to the appropriate quality standard. In such a case, the ICH guidelines on GCP should be taken appropriately into account for the application of the rules set out in this Regulation, provided that there is no other specific guidance issued by the Commission and that those guidelines are compatible with this Regulation.)
  • QP specifically interested in: Maintaining PSF, Appropriately packaging/ labelling for relevant markets, ensiring trial is being performed from an approved centre, Sponsor responsibilities are understood, responsibilities, communication lines/ agreement clear, clinical trial data integrity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is a comparator?

A

Eudralex vol 4. Annex 13:
An investigational or marketed product (i.e. active control), or placebo, used as a reference in a clinical trial.

If a product is modified, data should be available (e.g. stability, comparative dissolution, bioavailability) to demonstrate that these changes do not significantly alter the original quality characteristics of the product.
The expiry date stated for the comparator product in its original packaging might not be applicable to the product where it has been repackaged in a different container that may not offer equivalent protection, or be compatible with the product. A suitable use-by date, taking into account the nature of the product, the characteristics of the container and the storage conditions to which the article may be subjected, should be determined by or on behalf of the sponsor. Such a date should be justified and must not be later than the expiry date of the original package. There should be compatibility of expiry dating and clinical trial duration.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is the ‘Declaration of Helsinki’?

A

The World Medical Association (WMA) has developed the Declaration of Helsinki <in 1964> as a statement of ethical principles for medical research involving human subjects, including research on identifiable human material and data.

Principles related to clinical trials include:
“Medical research is subject to ethical standards that promote and ensure respect for all human subjects and protect their health and rights.”
“Medical research involving human subjects must be conducted only by individuals with the appropriate ethics and scientific education, training and qualifications.”

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What should be included in the assessment of each IMP batch for QP certification prior to release?

A

Eudralex vol 4. Annex 13 details requirements for batch assessment:
- GMP and MA
- Audits of manufacturer
- Docs to certify the Manufacturer is authorised fo rthe IMP
- batch records re: compliance with the PSF, the order, protocol and randomisation code. (deviations or planned changes completed)
- production conditions
- the validation status of facilities, processes and methods;
- examination of finished packs;
- test results
- stability reports;
- conditions of storage and shipment;

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is an ‘Investigators Brochure’?
would you expect to see in this?
What sections are particularly important to the QP?

A

Most current definition is in 536/2014:
Brochure = a compilation of the clinical and non-clinical data on the investigational medicinal product or products which are relevant to the study of the product or products in humans;

ICH E6(R1) Good Clinical Practice. Protocol contents:
7.3.1 Table of Contents
7.3.2 Summary (significant physical, chemical, pharmaceutical, pharmacological, toxicological, pharmacokinetic, metabolic, and clinical information available)
7.3.3 Introduction
7.3.4 Physical, Chemical, and Pharmaceutical Properties and Formulation
7.3.5 Nonclinical Studies Introduction: (results of all relevant nonclinical pharmacology, toxicology, harmacokinetic, and investigational product metabolism studies)
7.3.6 Effects in Humans Introduction:
7.3.7 Summary of Data and Guidance for the Investigator (overall discussion of the nonclinical and clinical data)

<underlined>
</underlined>

25
Q

What is a SUSAR?

A

SUSAR = Suspected Unexpected Serious Adverse Reaction
Reporting of SUSAR is mandatory for clinical investigators in the European Union. A report must be submitted to the national competent authority within 15 days of occurrence (7 days in case of death or life-threatening issue.

Following introduction of 536/2014, SUSAR’s will be reported via the Clinical Trial Eudravigilance database.

26
Q

Your company is taking a new product IMP. What considerations do you make before then during GMP manufacture?

A

Product introduction assessment. Look at:
Before: Licenses, H&S, Drug potency & cleanability, Technical processing & analytical capabilities & transfer activities, Facilities, Documentation, Training, Quality agreements,

For Manufacture: MIA (IMP), PSF, CTA & BMRs, Approved raw material sources with Cof A and TSE statements.

27
Q

What is meant by ‘green light’ in reference to clinical trials?

A

The QP has a legal responsibility as laid out in Article 13 of 2001/20/EC to ensure that the IMP has been manufactured in accordance with EU GMP and meets the conditions of the clinical trial authorisation and the product specification file (PSF). In certifying a batch against the PSF, investigational medicinal product dossier or the clinical trial authorisation (CTA) the QP is providing certification, which has been known previously as the technical green light.

Release of IMPs for use in a clinical trial should not occur until after the QP has certified the batch(s). Under Article 9 of 2001/20/EC, the sponsor may not start a clinical trial until the clinical trial authorisation has been granted for the trial and all conditions of the authorisation have been met; and an Ethics Committee positive opinion has been granted and each trial site has been approved. The process for ensuring that the appropriate approvals are in place has been known previously as the regulatory green light.

In practice, although the QP certification and regulatory approval processes may be run in parallel, the sponsor is responsible for ensuring both steps are completed prior to the release of IMPs for use in a clinical trial. The QP certification must be provided by a QP named on the MIAIMP licence specified in the clinical trials authorisation as responsible for the manufacturing and importation of the IMP. The regulatory green light release may be delegated by the sponsor to the QP, regulatory affairs or trial manager. However the sponsor retains legal responsibility

28
Q

What is meant by ‘green light’ in reference to clinical trials?

A

The QP has a legal responsibility as laid out in Article 13 of 2001/20/EC to ensure that the IMP has been manufactured in accordance with EU GMP and meets the conditions of the clinical trial authorisation and the product specification file (PSF). In certifying a batch against the PSF, investigational medicinal product dossier or the clinical trial authorisation (CTA) the QP is providing certification, which has been known previously as the technical green light.

Release of IMPs for use in a clinical trial should not occur until after the QP has certified the batch(s). Under Article 9 of 2001/20/EC, the sponsor may not start a clinical trial until the clinical trial authorisation has been granted for the trial and all conditions of the authorisation have been met; and an Ethics Committee positive opinion has been granted and each trial site has been approved. The process for ensuring that the appropriate approvals are in place has been known previously as the regulatory green light.

In practice, although the QP certification and regulatory approval processes may be run in parallel, the sponsor is responsible for ensuring both steps are completed prior to the release of IMPs for use in a clinical trial. The QP certification must be provided by a QP named on the MIAIMP licence specified in the clinical trials authorisation as responsible for the manufacturing and importation of the IMP. The regulatory green light release may be delegated by the sponsor to the QP, regulatory affairs or trial manager. However the sponsor retains legal responsibility

29
Q
  1. What are the differences between legal duties of commercial QP and IMP QP? Where would you find legal duties of IMP QP?
A

The differences
- Manufacture to MA compliance whereas IMP’s are to PSF and other relevant authorisations e.g. IMPD
- Importation to compliance with MA, quantitative for all API’s and qualitative for all other tests, all other tests comply with MA and accept on CofA if MRA in place. However for IMP’s, it must be made to a GMP equivalent to EU GMP.
- Testing carried out in compliance with PSF for IMP’s
- Affixing of safety features is not noted for legal duties for IMP QP.

30
Q
  1. What is 536/2014? From the new detailed commission guideline (new Annex 13) what are the principles UK decided to adopt?
A

IMP Regulation within Europe, however it came into force once Britain had left EU so does not apply to the UK.

31
Q
  1. What is the content of PSF and impd ?
A

PSF
- Specifications and methods
o SM, PM and intermediates, bulk and finished product
- Manufacture methods
- IPC testing and methods
- Approved label copy
- CT protocol and randomisation does
- Technical agreements
- Stability data
- Storage and shipment conditions
IMPD
- Quality data for
o drug substance (structure, manufacture, process controls, CCP’s, spec, ref standards and stability)
o Medicinal product (development, components, excipients, properties, micro attributes, mfr, mfr controls, CCPs, process validation, specifications, analytical procedures, impurities, ref standards, container closure system and stability
- Non-clinical pharmacology and toxicology data
- Previous CT and human experience data
- Overall risk and benefit assessment

32
Q
  1. What is double blinded study? What controls you would expect to see in primary packing of such type of trial? How would you ensure that blind is maintained throughout the trial? What documentation you would require?
A

Neither the patient not clinician know if the patient is taking the IMP. The primary packaging of IMP and the placebo/comparator must look identical. Randomisation codes are used and maintained by the sponsor, who can deduce if the patient is taking the treatment

33
Q
  1. How would you import an EU licensed product for a UK paediatric clinical trial (no UK license product available)?
A

Follow GDP practice and import into a WDA licenced site.

34
Q
  1. What are the legal duties of a commercial QP for human and veterinary products? Do veterinary products need safety features? What are the differences compared to an IMP QP?
A

Legal duties for commercial
- Manufacture and testing to EU GMP, compliance with MA and national laws
- Importation full qualitative analysis, quantitive analysis of actives and all other tests must comply with MA, for MRA countries, can accept on CofA.
- Certification documented in a register of equivalent document
- Safety features affixed to packaging

Legal duties for veterinary products
- Manufacture and testing to EU GMP, compliance with ManA and national laws
- Importation full qualitative analysis, quantitive analysis of actives and all other tests must comply with MA, for MRA countries, can accept on CofA.
- Certification documented in a control report
- Safety features not required for veterinary products.

Differences to IMP QP duties
- Manufacture to compliance with PSF and other relevant authorisations e.g. IMPD
- Importation must be manufactured to a GMP equivalent to EU GMP.
- Testing carried out in compliance with PSF for IMP
- Affixing of safety features is not noted for legal duties for IMP QP.

35
Q
  1. You are a contract QP for a new client that will be performing FIH trials. What do you need in place before you start? What is required in a FIH trial? How do you determine dosing? The product will be in a vial / infusion bag with saline. What are your concerns regarding formulation?
A

(Phase 1) First in human trials is a small number of healthy volunteers to determine safety profile of drug. IMPD, CT Protocol, PSF, ethics committee approval, manufacturing process and any defects and deviations noted.

Dosing is determined by protocol, dosage regime listed (single dose, titrating dose or escalation)
Sterility and reconstitution of product, any other equipment needs to be provided, vials and temp + storage

36
Q
  1. (What are the legal duties of the QP and where do you find them?) What would be different for an IMP QP?
A

(Legal duties for commercial
- Manufacture and testing to EU GMP, compliance with MA and national laws
- Importation full qualitative analysis, quantitive analysis of actives and all other tests must comply with MA, for MRA countries, can accept on CofA.
- Certification documented in a register of equivalent document
- Safety features affixed to packaging
For EU – 2001/83 Article 51
For UK – Human Med Regs 2012:1916 Schedule 7 Part 3)

Differences to IMP QP duties
- Manufacture to compliance with PSF and other relevant authorisations e.g. IMPD
- Importation must be manufactured to a GMP equivalent to EU GMP.
- Testing carried out in compliance with PSF for IMP
- Affixing of safety features is not noted for legal duties for IMP QP.
For EU – 536/2014 and 2017/1569
For UK – SI2004:1031 Part 6 Section 43

37
Q
  1. IMP scenario – Product is imported to UK from India, Secondary packed in UK. What do you need to be in place from a QP perspective to certify?
A

Technical agreement with manufacturer in India, audit of India site to ensure that manufacturing is equivalent to UK standards.

Import testing in the UK once the product has been received.

Ensure secondary packaging site in UK has MIA(IMP). TA in place

Agreement and responsibilities defined between manufacturer, packaging site, sponsor, QP release site.

Reference and retain samples, QP decleration (for IMP’s)

38
Q
  1. Your company now wants to change from an Indian manufacture to manufacture and all packing activities in Germany. What do you need to do? How is the QP role changed?
A

Change control raised, types of products and impact assessment to be conducted. Tech transfer, testing transfer to German site. Update IMPD and PSF with latest manufacture, packing and testing details.
Assessment/audit of the German manufacturing and packing site. Ensure MIA(IMP) in place.
UK QP no longer doing IMP certification but ensuring that the QMS is fit for purpose.
Receive product into UK Hub or investigator site. Verified it has been QP released by a UK QP.

39
Q
  1. What is the term for a clinical trial where you have an active and a placebo tablet but now want to introduce a liquid comparator?
A
40
Q
  1. Scenario – in a Ph3 clinical trial running in France and Germany (NI QP) the French site is struggling to recruit and the sponsor wants to move packs from the German site which isn’t having much success recruiting. What do you think?
A

Will packs be in German or French? Are the randomisation codes not based on the number of patients recruited,
Blocks and move entire block to remove issues
Labelling requirements

41
Q
  1. Scenario – When you started the trial you had a 9-month shelf life on the product. That can now be extended with data to 18 months. Can the packs on the market be updated?
A

Yes the shelf life can be extended, this is preferentially carried out at a MIA(IMP) site that is certified for repackaging or can be carried out at the clinician site. The packs must be brought back to the site by the patient to have the shelf life extended. This process does not require re-QP release of the packs.

42
Q
  1. Whose responsibility is it to perform the update?
A

Sponsor

43
Q
  1. What data needs to be included on the updated label?
A

New expiry, batch number, randomisation code, CT reference number.

44
Q
  1. Do you know about blinding of IMPs when packaging.
    a. How would you control this?

b. What type of trial would you run to compare an IMP and comparator?

c. How would you measure therapeutic effect of IMP compared to comparator?

d. What are the considerations for getting the comparator from the US?

e. Your supplier for a multicentre trial has been sourcing comparator from the EU as opposed to the US, what would you do?

f. What else would you need to change (I clarified they meant a substantial amendment)

A

a. The packaging regardless of if the IMP or the placebo/comparator looks the same.
b.
c. Compare against the market leader/ bench mark against it, compare IMP result against what is currently approved, does it show improved efficacy/therapeutic effect or reduced side effects.

d. May not have a marketing authorisation in the UK, treat the same as an IMP, so testing to be carried out and QP released. Manipulations of the comparator – effect on shelf life?

e. Raise a deviation as the details within the IMPD and PSF were not correct. Assess difference in the EU and the US product, and carry out full impact analysis. May not need QP release as it will have been released by EU QP, but oversight of QMS from UK QP.

f. IMPD and PSF and all other documents registered with the regulatory body.

45
Q
  1. Where do you find the legal duties for a commercial and IMP QP? What are the legal duties for both? You mentioned 3rd country what do you mean by that?
A

(Legal duties for commercial
- Manufacture and testing to EU GMP, compliance with MA and national laws
- Importation full qualitative analysis, quantitive analysis of actives and all other tests must comply with MA, for MRA countries, can accept on CofA.
- Certification documented in a register of equivalent document
- Safety features affixed to packaging
For EU – 2001/83 Article 51
For UK – Human Med Regs 2012:1916 Schedule 7 Part 3)

Differences to IMP QP duties
- Manufacture to compliance with PSF and other relevant authorisations e.g. IMPD
- Importation must be manufactured to a GMP equivalent to EU GMP.
- Testing carried out in compliance with PSF for IMP
- Affixing of safety features is not noted for legal duties for IMP QP.
For EU – 536/2014 and 2017/1569
For UK – SI2004:1031 Part 6 Section 43

3rd Country is a country or territory in which the members of the EU can move freely between, or have an MRA in place with.

46
Q
  1. You have a blinded IMP study – what is meant by blinded? What would you do if your IMP was a capsule, and your comparator was a little white tablet?
A

Blinded means the patient is not aware if they are taking the IMP or the comparator/placebo. Single blinded is if the clinician knows and double blinded is if neither clinician or patient knows.
The white comparator tablet can be over encapsulated into the same capsule shell as the IMP product.

47
Q
  1. You have a clinical trial for oncology – blinded study – you find out that the comparator has been sourced from a non-bonafide source behind your back, what are your concerns – you have no idea over quality
    a. Directors are pressurising you as its oncology and the patient needs it – what do you do?
A
48
Q
  1. What is in a PSF and CTA
A

PSF
- Specifications and methods
o SM, PM and intermediates, bulk and finished product
- Manufacture methods
- IPC testing and methods
- Approved label copy
- CT protocol and randomisation does
- Technical agreements
- Stability data
- Storage and shipment conditions

CTA
-Trial ID
- Sponsor Information
- Info on each IMP
- Trial info
- subject population
- Trial sites
- Ethics committee

49
Q
  1. Statin label requires update for a minor safety update
    a. How would you manage this process
    b. What if this was an IMP product how would the process differ. This lead into what other types of activities can be performed at CT site (talked about expiry updates.
    c. Does this always have to be done under an MIA, does it always have to be CT pharmacist if being done at the site, (no talked about training other personnel)
    d. How would you manage a repackaging process if required.
A
50
Q
  1. Talk me through different CT design approaches. I was stopped shortly after mentioning open study and we moved on to another question.
A
51
Q
  1. What are the phases of clinical trials? At each phase, could you give a brief description/overview
A
52
Q
  1. Which part of the CTD is of interest to the QP and why? (Ref Module 3)
    a. Which other part is also important to QPs and why? (Ref Module 1)
    b. Where will you find guidance? (ICH M4, Dir 2001/83EC; Eudralex)
A
53
Q

Scenario, you are a QP on an IMPs site and involved in double blind trial. You have 10mg, 100mg and placebo tablets in blisters. 10mg and placebo tablets are same size and same colour whereas 100mg tablets are different colour and bigger in size. As a QP what are your considerations and concerns to ensure that the trial runs successfully?

A
54
Q

Can you sketch a packaging line for IMPs and your considerations in terms of in‐process checks in comparison to a packaging line for commercial products?

A
55
Q

Packaging on IMPs (single sterile vial in carton). What controls would you see around the packaging operation for i) an open label study and ii) a double blind study? What would you expect to see on the label text for i) an open label and ii) a double blind study? What would you expect to see on the label for a commercial product and where is this information found?

A
56
Q

Your commercial product site has decided to start making IMPs. They are making IMPs for a double blind placebo study. What needs to be considered for this major change?

A
57
Q

You receive a complaint for a product which is part of a double blind clinical trial study. The complaint relates to the incorrect dosage information on the secondary packaging. The product is a sterile injectable. What do you?

Where would you find the requirements of the printed component?

What is the content of the CTA? Where within the CTA would you find details of the printed component?

A
58
Q

What would you expect to see on an IMP label?

Are there any labelling changes that a QP needs to be aware of as a result of the
new CTR?

A

Name, drug, strength, form
route of admin, quantity
CT refernece number
Subject ID number
Investigator
Directions for use
‘For clinical use only’
Storage conditions
Period of use (expiry/retest date)
additional warning symbols
‘out of reach of children’

59
Q

IMP currently has a 12‐month shelf life and batch is 11 months old. Now have stability data to support 18‐month shelf life. Shelf life extension has been approved. Stock in your warehouse and at clinical trial sites.

Can you extend the shelf life of the IMP?

How would you do this for material in your warehouse and at clinical trial site?

Shelf life extension only applies to IMP and not placebo or compactors. Your study is blinded. What do you do now?

A