Sei sulla pagina 1di 107

Good Practices for demonstrating safety and

Commented [a1]:
quality through recipient follow-up Audit Comment:

significant part of the guide is


to help establish whether a TCTP is
novel or not, yet there is no
mention of it in the title. I
appreciate the title comes from the
title of the grant. The title mentions
recipient follow up yet there is no
reference to it in the title of the
guide.

Guidance, Methodologies and Tools –


(2nd Draft –7th March 2018)

This guide] is part of the project‘ 709567 / Euro-GTP II’ which has received funding from the European Union’s
Health Programme (2014-2020).”
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Collaborations: ...................................................................................................................................... 4
Acronyms: .............................................................................................................................................. 5
1. Introduction ............................................................................................................................... 6
1.1. Purpose of Euro GTP II Guide: ........................................................................................... 7
1.2. Project Rationale and Objectives ........................................................................................... 8
1.3. Structure of this document .................................................................................................... 9
1.4. How should the guide be used? ............................................................................................. 9
1.5. Ethical aspects ...................................................................................................................... 10
2. Methodology for TCTP characterization, assessment of novelty and risk evaluation ................ 11
2.1 Characterization of the TCTP ................................................................................................. 11
2.2. Evaluation of Novelty (Step 1) ............................................................................................ 11
2.3. Overview of the risk assessment process – Level risk analysis (Step 2) ............................... 13
2.4. Definitions of Studies Extent (Step 3):.................................................................................. 16
3. Instructions for the correct use of EuroGTP II methodologies and tools .................................... 20
3.1. Accessing the IAT ................................................................................................................. 20
3.2. Key principles for effective use of the EuroGTP II methodologies and IAT .......................... 20
3.3. When should the IAT be used? ............................................................................................ 21
3.4. Step 1 : Evaluation of novelty ............................................................................................... 21
3.5. Step 2: Level risk analysis - Identification and Quantification of Risk .................................. 22
3.6. Step 3: Definition of extent of studies needed based on the risks quantified ..................... 27
4 . Tissues specific chapter .............................................................................................................. 29
4.1. Evaluation of Novelty (Step 1) .............................................................................................. 29
4.2. Level risk analysis (Step 2) .................................................................................................... 31
4.3. Definition of extent of studies needed based on the risks quantified (Step 3) ................... 40
4.4. Worked examples ................................................................................................................. 48
5. Hematopoietic Stem Cell Specific Chapter .................................................................................. 49
5.1. Evaluation of Novelty (Step 1) .............................................................................................. 49
5.2. Level risk analysis (Step 2) .................................................................................................... 52
5.3. Definition of extent of studies needed based on the risks quantified (Step 3) ................... 59
5.4. Worked examples ................................................................................................................. 68
6. ART Specific Section .................................................................................................................... 69
6.1. Evaluation of Novelty (Step 1) .............................................................................................. 69

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


2
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

6.2. Level risk analysis (Step 2) .................................................................................................... 71


6.3. Definition of extent of studies needed based on the risks quantified (Step 3) ................... 82
6.4. Worked examples ................................................................................................................. 92
7. Tissue & Cell Database ................................................................................................................ 93
7.1. Purpose of the T&C Database .............................................................................................. 93
7.2. General Principles ................................................................................................................. 94
7.3. Introduction of data: ............................................................................................................ 94
7.4. Description of Contents ........................................................................................................ 95
7.5. Codes used: .......................................................................................................................... 97
7.6. Structure of Data .................................................................................................................. 97
7.7. Queries and Practical Use of the database: ......................................................................... 98
7.8. Management of Contents and Certification Procedures ..................................................... 98
Bibliografía......................................................................................................................................... 100
Annex I – Partners and Experts of EuroGTP II Project ....................................................................... 103
Annex II – Template Form: Characterization of TCTP ....................................................................... 106
Annex III – Template form: Methodologies for Assessing the Risks associated to novel TCTP ....... 107

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


3
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Collaborations:
EuroGTP II project is strictly connected with:

 VISTART Joint Action - meant to support EU Member States (MS) in developing and
strengthening their capacity for monitoring and control quality, safety and efficacy in
the field of blood, tissues and cells transplantation.
 ECCTR Project - aims to build a common assessment methodology and establish an
EU web-based registry and network for academics, health professionals and
authorities to assess and verify the safety quality and efficacy of corneal
transplantation.
Collaboration with this project with is considered worthwhile, as the use of registries
is considered an important tool for the evaluation of efficacy and safety of SoHO. The
criteria identified by ECCTR are also considered to be a valuable example for the
definition of follow-up and clinical evaluation principles by EuroGTP II project.
 GAPP Action (facilitatinG the Authorisation of Preparation Process for blood and
tissues and cells) JA – having in mind the need for future requirements associated
with the clinical evaluation of efficacy and safety performed by NCAs, and the links
needed to assure the coherence between EuroGTP II outcomes and any future tools
developed, the Coordinator (BST) requested the participation as Associative Partner
in the future JA.

These collaborations aim to develop harmonized procedures and Good Tissue and cells Practices
(GTPs), for the different European stakeholders: Tissue Establishments (TE), Organisations
Responsible for Human Application (ORHA), and National Competent Authorities (NCA).

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


4
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Acronyms:
ART – Assisted Reproductive Technologies
ATMP – Advanced-Therapy Medicinal Product
CA – Competent Authority
CA – National Competent Authority
CBB – Cord Blood Bank
CHAFEA – Consumers, Health, Agriculture and Food Executive Agency
CoE – Council of Europe
CPPs – Critical Process Parameters
CQAs – Critical Quality Attributes
EC – European Commission
ECCTR – European Cornea and Cell Transplantation Registry
EDQM - European Directorate for the Quality of Medicines
ESSKA - European Society for Sports Traumatology, Knee Surgery and Arthroscopy
EUTCD – European Tissue and Cells Directives
GAPP – facilitatinG the Authorisation of Preparation Process for blood and tissues and cells
GTP – Good Tissue and Cells Practices
HPC – Haematopoietic Progenitors Cells
HSC – Hematopoietic Stem Cells
IAT – Interactive assessment tool
IVF – In Vitro Fertilization
MS – Milestones
ORHA – Organisation Responsible for Human Application
QC – Quality Control
SARE – Serious Adverse Reactions and Events
SoHO – Substances of Human Origin
T&C – Tissues and Cells
TCTP – Tissue and Cellular Therapy/Product
TE – Tissue Establishment
V&S - Vigilance and Surveillance
VAS - Visual Analogue Scale
VISTART – Vigilance and Inspection for the Safety of Transfusion, Assisted Reproduction and
Transplantation
WP – Work package

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


5
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

1. Introduction
Preparation of Tissue and Cellular Therapies and Products (TCTPs), including reproductive cells,
intended for human applications should comply with high standards of quality and safety according
to the requirements established by the EUTCDs [1] [2] [3] [4] [5] [6], in order to ensure a high level of
health protection in the Community. This concept becomes even more important with new products
that are applied for the first time in humans or are prepared with new and innovative methodologies.
Advances in basic science, technology and medicine continually create opportunities for new and
improved TCTPs. These may be wholly new types of TCTPs, or improved methodologies for the
preparation of existing TCTPs. While the objective of these changes and novelties is to prepare TCTPs
that are safer, clinically more effective and adequate to the needs of clinicians and patients, there is
always the risk that any change in the processing method can cause a potential harm in the recipient.
It is therefore vital that an evaluation of the potential risk of a process is implemented whenever a
significant change is made.
To date, no regulations nor standardized methodologies have been established, however with a
recent VISTART document, Competent Authorities (CAs) of Tissues and Cells (T&C) have introduced
the first principles about this topic. This represents the basis of a future regulatory framework based
around the need to gather clinical follow up of recipients as a means of validation of TCTPs prepared
with newly developed processing methodologies. Furthermore safety and efficacy studies, based on
risk based assessment are recognized to be needed as has been indicated by the EU results of the
survey carried out by the EuroGTPII project
Being aware of the importance to improve standards for quality, safety and efficacy of TCTPs,
especially those related to innovative TCTPs, the European Commission has funded the EuroGTP II
project (European Good Tissue and cells Practices II) - “Good Practices for demonstrating safety and
quality through recipient follow-up”. The main objective is to set up good practices with regard to
pre-clinical and clinical evaluation of for human Tissues, Haematopoietic Stem Cells (HSC) and
Assisted Reproduction Technologies (ART) therapies and products. Euro-GTP II will give continuity to
the first EuroGTP project [7], which has developed European Good Tissue Practices for the activities
carried out in TEs.
By using the a systematic approach proposed in the current guide users will be able to evaluate risks
resulting from the donation, preparation, and clinical application processes and to design
appropriate studies (pre-clinical, clinical and clinical follow up programs). These should be
proportionate to the level of residual/unknown risk after the validation procedures and contribute to
confirm that the TCTP is safe and effective.
Good practices, principles and reference tools applicable to TCTPs and how to conduct adequate
clinical follow up studies identified by this document will represent a useful starting point to facilitate
this common approach.
The proposed methodologies aim to be systematic and consistent, in order to promote the
harmonization of practices and recognition amongst the EU stakeholders, namely, TEs, ORHAs and
CAs.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


6
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

The definition of the “novelty threshold “ of a change, the methodology to identify and measure the
risks connected to the new process, and defining the extent of preclinical and clinical studies to be
submitted to the CA for the new PPA are the main topics described in this document. Commented [a2]: We are not focus on the PPA

The current good practices should be seen as a standard for the design, conduct, performance, VISTART and GAPP are tools for CAs, but we are developing tools
for TEs and ORHAs (that can be used also to provide information to
monitoring, auditing and recording of the studies needed to assess, prove and document efficacy and CAs.)
safety of TCTP.
This paragraph can be deleted in this section

1.1. Purpose of Euro GTP II Guide:


The purpose of this document is to provide structured guidance on how to use the tools and
methodologies made available by the EuroGTP II project.
This guide has been elaborated with the collaboration of experts and representatives of EU TEs,
OHRAs, scientific associations, universities, CAs, research organizations, and national registries
(Partners and Experts of EuroGTP II Project - Annex I).
In order to ensure alignment and coherence among existing documents dealing with patient follow –
up programs the above mentioned stakeholders have taken into account the following current
guidelines and reference documents:

 VISTART deliverable about regulatory principles for clinical follow up of recipients (Draft
Principles for Competent Authorities for the Evaluation And Approval of Clinical Follow Up
Protocols For Blood, Tissues and cells Prepared with Newly Developed And Validated
Processing Methodologies);
 ARTHIQS (Assisted Reproductive Technologies and Haematopoietic stem cells Improvements
for Quality and Safety throughout Europe)recommendations for cord blood banks [8] and
ARTHIQS about donor follow up registries [9];
 Outcomes of SoHO Vigilance & Surveillance (V&S) Project [10]
 Deliverables of European Union Standards and Training for the Inspection of Tissues
Establishments (Eustite) Project [11]
 Deliverables of European Good Tissue Practices (EuroGTP) Project [7]
 Guide to the Quality and Safety of Tissues and Cells for Human Application, 2017, 3rd edition,
Council of Europe (CoE), European Directorate for the Quality of Medicines (EDQM) [12]
 The Single European Code for Tissues and Cells (SEC) - Reference Compendia for the
Application of a single European Coding System for Tissues and Cells
 Notify Library – Global Vigilance and Surveillance Database for Medical Products of Human
Origin
 FACT-JACIE (Joint Accreditation Committee - ISCT & EBMT) International Standards for
Hematopoietic Cellular Therapy Product Collection, Processing, and Administration [13]
 ESHRE (European Society of Human Reproduction and Embryology) guidelines for good
practice in IVF laboratories [14]
The EuroGTP II established the structure of the Tissue & Cells Database (Chapter 7), since the
information about quality and safety aspects of the T&C processes and the clinical outcome of the
recipient gathered by the TEs is representing a valuable patrimony of scientific and technical data it
would be beneficial to the interested stakeholders to share this knowledge.
Finally the EuroGTPII will be used as a basis for the next Joint Action GAPP that will develop the
criteria for the quality aspect of preparation processes.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


7
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

1.2. Project Rationale and Objectives


There are three key outputs from the Euro GTP II Project:
A. To develop a systematic, risk based mechanism and Interactive Assessment Tool (IAT) to: Commented [TK3]: Maybe to add:
 Implement the results of risk assesment into the daily practice
Evaluate if a new or changed TCTP has significant novelty and follow-up the results
I think this tool is important to became a paramount requirement ,
 Determine the overall risk arising from the novelty not only from a clinical but also from an ethical point of view. Risk
assesment on novelty is crucial to ensure maximum safety for
 Determine an appropriate level of pre-clinical and clinical evaluation to address the risk patients. (IR)

Chapter 2 provides a more detailed methodology for this objective.

Figure 1 – Core Euro GTP II Process

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


8
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

B. To create a Tissue and Cells (T&C) database of tissues/cells products, preparation processes,
applications and therapies: Commented [TK4]: Also , I think some data on the incidence of
short and long-term adverse reactions must be added into database
 The purpose of this database will be to promote the safe use of TCTPs, by the provision of cfr. Notify. (IR)

data related with the products and therapies available, and their respective efficacy
references.
The structure and contents of the database were defined in order to assure its consistency,
harmonize the characterization of TCTPs, and support the collection of efficacy and quality
data associated with the clinical use of Substances of Human Origin (SoHO) at European
level.
More details can be found in the Chapter 7 of this document (add future link)

C. To put in place mechanisms to ensure the sustainability of the project’s outcomes and propose a
structure for the development of European accreditation and training programmes for TEs, ART
centres and ORHAs:

 This model aims to assure the continuity and sustainability of the outcomes of the Euro-GTP
II Project, and the future update, promotion and harmonization of GTP’s standards. Commented [TK5]: Harmonised procedures also at national
level with shared reference documents and tools results. There
(we can add the future link for this deliverable) should be a text regarding the implementation on the local national
level - is this going to be the job of the local transplantation
authority? Is the implementation going to be obligatory? Penalty
for failiing to comply? (ZV)
1.3. Structure of this document
The guide is structured in 7 principle chapters:
Chapter 1 – General Introduction and overview
Chapter 2 – Methodology for TCTP characterization, assessment of novelty and risk
evaluation
Chapter 3 – Instructions for the correct use of EuroGTP II methodologies and tools.
Chapter 4 – Specific guidance with regard to using the IAT for tissue products and therapies
Chapter 5 – Specific guidance with regard to using the IAT for stem cell products and
therapies
Chapter 6 – Specific guidance with regard to using the IAT for assisted reproduction
technology (ART) products and therapies
Chapter 7 – A guide to the structure and use of the T&C database

1.4. How should the guide be used?


It is suggested that chapters 1,2 and 3 of this guide be read in their entirety before attempting to use
the methodologies proposed by the EuroGTP II Project.
Chapters 4, 5 and 6 are intended to be used as reference and support when using the IAT, as they
provide specific guidance for the use of tools and methodologies applied to the different areas of
SoHO.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


9
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

1.5. Ethical aspects


New insights are leading to the development of different (innovative) treatment protocols [15], and
may succeed in the introduction of a novelty tissue or cellular therapy/product (TCTP) into clinical
practice.
The application of a novelty may be considered an experimental treatment, and as such, it is required
to meet the principles of medical ethics. Experimental therapy is often the place where scientific
research and clinical practice meet. Understanding and application of basic ethic principles
(autonomy, non-maleficence, beneficence and justice) is essential for the clinical approach of novelty
treatments. Institutional Review Boards or Medical Ethics Committees review research proposals,
including the (impact of ) procurement, source of tissues, and expected results in order to assess
whether ethical principles are met. It is part of the process to justify safeguarding the recipient and
donor, and confirm moral correctness.
If one looks from a patient’s perspective, there are other ethical aspects that touch on the patient’s
well-being, and personal interests should not be underestimated. Giving informed consent to an
experimental treatment means the patient’s role switches into the role of volunteer. Principles as
dignity, integrity and vulnerability become more apparent. [16].
One can argue that patients, for whom no alternative evidence based treatment is available, have a
‘right-to-try’ medical therapies for which no evidence based proof is currently presented [17],
ignoring any potential risks that the treatment carries with it [18].
Although health care is a fundamental right, it is known that access to therapies is not for all patients
reality. Thorough studies to validate novelty TCTP are virtually impossible, due to
• the scarcity of donated bodily material (is it ethically justified to use 5-
10% of the donated TCTP for validation tests?)
• the lack of preclinical studies (using animal settings,
• the lack of randomised clinical trials. Donated TCTP differ from donor to
donor, and also the recipients of the novelty products are each unique.
Concerns raise about the access to products, the therapeutic value and the impact of the use not
only on recipient but also on the society [19]. Resources for health care are limited, insufficient to
meet the needs of all patients, and forces into making choices. Access to novelties or new therapies
is often restricted to so-called ‘compassionate use’. Initiatives, both from patient-driven and
commercial organisations, are developed to increase access to and availability of TCTP for rare
diseases [20] [21]. In that light, optimistic (premature) publicity, or an appeal to novelty, might give
false hope to patients, resulting in fatalities. [22]
Even if preliminary results of a novelty TCTP are promising, the translation from research to the
delivery of the innovative stem-cell based practice is often delayed due to a variety of (European)
regulations [23], and the lack of resources. In light of the latter, patients with rare diseases, patients
living in small or less rich countries, may less benefit from novelty TCTP [24].

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


10
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

2. Methodology for TCTP characterization, assessment of novelty and risk evaluation

The assessment methodologies proposed by the EuroGTP II project can be used using the
available templates (Annex III) or the IAT.
In any case, instructions related with the correct use of these methodologies can be found in
the chapter 3 and/or in the SoHO specific chapters (4, 5, and 6) of this guide.

2.1 Characterization of the TCTP


Before commencing assessment of novelty and risk, it important that the TCTP is thoroughly
evaluated and characterised so that the process can be performed accurately. This requires that the
following details be documented (proposed template – Annex II):
 Justification for the implementation of change, including the key benefits of the innovation.

 How is the TCTP prepared; what, if any, changes have been made to the preparation or
treatment protocol?
 What is the origin of the TCTP (autologous or allogeneic or in case of ART concerning partner
or non-partner donation)?
 In what form is it presented for clinical application?
 What, if any, excipients or other reagents or residues could be transferred upon clinical use
with the TCTP (such as carriers or preservants)?
 What are the critical process parameters applied to the TCTP preparation protocol?
 What are the critical quality attributes necessary for the TCTP to deliver its intended result?
 What clinical indication is the TCTP to be used for?

Additionally, prior to the implementation of changes/new processes a formal document detailing the
factors that justify such developments should be produced. This document should describe:
 Existence of previous clinical data reported by other centres (if applicable);
 Means of control and additional quality indicators evaluated;
 Overview of the clinical intended action of the TCTP;
 Bibliographic evidences that support the implementation of changes.

2.2. Evaluation of Novelty (Step 1)


It is important that the definition of ‘novelty’ within the context of this process is clearly established
and understood. It is not intended to encompass every change to a product or process, regardless of
how minute the change is; rather it intends to capture any change that could significantly affect the
quality and/or safety of the TCTP and/or the safety of recipients. This is the first step of the novelty
and risk evaluation process (Figure 2 below)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


11
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Figure 2 – Evaluation of Novelty - This assessment involves answering a series of seven questions,
covering all aspects of TCTP provision from donor selection to clinical application of the final product.
If no novelty is identified, it can be concluded that there is no significant change or innovation in the
TCTP being assessed. (This process is discussed in detail in the Chapters 3, 4, 5 and 6.)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


12
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

2.3. Overview of the risk assessment process – Level risk analysis (Step 2)
Having established that a new or changed TCTP has significant novelty, a systematic risk assessment
must be undertaken to identify and quantify the risks associated with it. This must be a
comprehensive process that considers all aspects of TCTP provision, from donor selection through to
implantation or application of the product or therapy. This is the second step of the novelty and risk
evaluation process (Figure 3 below)

Figure 3 – The risk assessment process- The risk profile is determined through the identification of
potential risks factors ( Figure 4) and analysis of risks (Figure 5). This is further explained in Chapter 3,
and in subsequent specific chapters relating to Tissues, Cells and ART with some examples.
Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015
13
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Figure 4: Risk factors

Figure 5: Potential Risks

The overall process requires that firstly, specific risks relating to the potential risk factors (Figure 4)
and risks (Figure 5) be identified. Each of these must be individually risk assessed to determine the
residual risk of implementing the change, assessed by considering
i) The probability of the risk occurring
ii) The severity of the consequences should the risk occur
iii) The probability that, should the risk occur, it will be detected
iv) Any existing evidences that can be used to mitigate the risk

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


14
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

The outcome of this exercise will be a single, overall risk score that can be used to determine the
definition and extent of pre-clinical and clinical studies necessary to support the proposed novelty or
change.
The tool used to quantify risk (described in detail in the next chapter) takes into account the number
of individual risks assessed when calculating the proportional risk score. Thus a process where
multiple minor risks are identified could generate the same overall risk score as process where a
single major risk is identified.
The quantity and quality of the available evidence, such as published data in peer reviewed literature
and internal validation reports, can be used to reduce this overall risk score. The whole risk
assessment process is explained in more detail in Chapter 3

EuroGTP II Algorithm in a nutshell:


(Demonstration of the algorithm with practical example - Annex xxx)
1. Estimate the risk value associated with the TCTP:

𝑅𝑖𝑠𝑘 𝑉𝑎𝑙𝑢𝑒 = Ʃ 𝑟𝑖𝑠𝑘𝑠 =


= Ʃ (( 𝑆 × 𝑃 × 𝐷) − (( 𝑆 × 𝑃 × 𝐷) × (%𝑟𝑖𝑠𝑘 𝑟𝑒𝑑𝑢𝑐𝑡𝑖𝑜𝑛))
P = Probability
S = Severity
D = Detectability

The proportional risk score classification is determined following the described steps:

𝑅𝑖𝑠𝑘 𝑉𝑎𝑙𝑢𝑒 × 𝐻𝑖𝑔ℎ𝑒𝑠𝑡 𝑃𝑜𝑠𝑠𝑖𝑏𝑙𝑒 𝑅𝑖𝑠𝑘 𝑠𝑐𝑜𝑟𝑒


𝑃𝑟𝑜𝑝𝑜𝑟𝑡𝑖𝑜𝑛𝑎𝑙 𝑅𝑖𝑠𝑘 𝑆𝑐𝑜𝑟𝑒 =
(𝑀𝑎𝑥 𝑆 × 𝑀𝑎𝑥 𝑃 ×𝑀𝑎𝑥 𝐷 ×𝑁𝑢𝑚𝑏𝑒𝑟 𝑜𝑓 𝐴𝑝𝑝𝑙𝑖𝑐𝑎𝑏𝑙𝑒 𝑅𝑖𝑠𝑘𝑠)

Max P = 5
Max S = 4
Max D =5
Applicable Number of Risks = Range from: 1 to 50 for tissues (including gonadic tissues); 1 to 45 for
HSC; 1 to 32 for ART (See details in the specific chapters 4, 5, 6)
Highest Possible Risk Score = (Max S × Max P ×Max D ×Number of Risks) x Risk Factors = 5000 for
Tissues; 4500 for HSC, and 3200 for ART

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


15
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 2.1. Overall risk classification


0 – 25 Negligible Risk
2 6-250 Low Risk
250 – 999 Moderate Risk
>1000 High Risk

The risk value doesn’t has a direct correspondence with the overall risk classification.

The calculation of score for risk classification needs to me proportional to the number of risks
evaluated in the assessment of the TCTP.

2.4. Definitions of Studies Extent (Step 3):


To Be Completed ... incl. Pre clinical, clinical studies and follow up

The value of the risk score generated by the risk assessment process determines the proportional
extent of studies required to ensure the safety and efficacy of the TCTP or therapies, in terms of the
pre-clinical (in vitro and/or in vivo) and clinical assessment. The specific, individual risks identified
further inform the precise test criteria indicated. The methodology proposed (in the chapters 3, 4, 5
and 6) in this guide will assist users in defining these protocols.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


16
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Figure 6 – The risk reduction and consequent definition of studies extent will attend all the risk factors
in a proportional relation to the risk values. The extension of studies needed will be proportional to
the level of novelty and level of risk associated.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


17
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 2.2. Extent of studies according the level of risk determined in the assessment

Level of Risk Proposed studies extent

The assessment indicates that the TCTP is safe and efficacious for clinical use and
unlikely to cause harm to recipients, however, it may be advisable to conduct a
revalidation of the process, if not already done. If the nature of the risk is not related
Negligible: to the process itself, the requirement for revalidation may not apply, for example
where the novelty is in the method of clinical application. No clinical follow up over
and above what is the mandatory requirement, such as serious adverse reaction and
event (SARE) reporting, is indicated.

The assessment indicates that the TCTP is safe and efficacious for clinical use and
unlikely to cause harm to recipients, however, a revalidation of the process, if not
already done, should be performed. If the nature of the risk is not related to the
process itself, the requirement for revalidation may not apply, for example where the
Low novelty is in the method of clinical application. In addition to the mandatory
requirement, such as serious adverse reaction and event (SARE) reporting, feedback
from immediate post transplant monitoring may be collected for a defined period or
number of procedures. Clinical audit may also be used after an appropriate period of
use.

The assessment indicates that more clinical evidence is needed to support safe and
effective use of this TCTP and mitigate risk. Process validation should be performed,
however if the nature of the risk is not related to the process itself, the requirement
Moderate for revalidation may not apply, for example where the novelty is in the method of
clinical application. Active collection of data on the safety and efficacy of the TCTP
should be put in place, until the risks have been adequately mitigated. Please refer to
chapter XX for additional details. Commented [a6]: Still needs major review..

The assessment indicates that significantly more clinical evidence is needed to


support safe and effective use of this TCTP and mitigate risk. Process validation
should be performed, however if the nature of the risk is not related to the process
itself, the requirement for revalidation may not apply, for example where the novelty
High is in the method of clinical application. Pre-clinical in vivo evaluation using an animal
model should be done if applicable (and if not already completed). The TCTP should
only be used clinically in the context of an ethically approved, controlled clinical
evaluation, until the risks have been adequately mitigated. Please refer to chapter XX
for additional details.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


18
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Design clinical evaluations adapted to T&C products/therapies


The design of clinical evaluation programs must be perform in close cooperation with the clinicians
responsible for the clinical application of the TCTP. The collaboration between TEs and end users is
critical to identify suitable design, clinical indication, number of patients, type of follow up
proportionate to the residual risks identified and ensure that comprehensive data is gathered to
evaluate efficacy.

Specific guidance relating to design of clinical evaluation for different types of TCTP will be provided
in Chapters 4-6. However, certain design considerations are common to all types of TCTP. There are
fundamentally two types of evaluation;
i) A study without control group (case series/registry approach), where the TCTP is used in
isolation
ii) A controlled study, where the TCTP is directly compared to a control treatment

The type of study selected will depend on a number of considerations, specifically:


a) The level of risk; if risk is high, a controlled study is more suitable, provided that it is feasible
for the TCTP in question.
b) The availability of a suitable control treatment.
c) The length of time that patients need to be followed up for; if long term follow up is
required, a controlled study may not be practical.

Moreover, the design of the clinical evaluation should consider:


a) The nature of the risk; if sudden graft failure is a significant risk, then patient recruitment to
the study should be graduated
b) The specific risks identified by the IAT, with assessment criteria to be identified with end
users.
c) The number of patients required to obtain statistically significant data

In addition to these considerations, TEs should endeavour to collaborate with fellow TEs to set up
multicentre studies, to ensure that sufficient patients are recruited. Collaborations with clinical trials
units should also be pursued to ensure that the requisite skills and resources are available to manage
studies.
Where feasible, a control group should be included in the clinical evaluation.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


19
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

3. Instructions for the correct use of EuroGTP II methodologies and tools


There are three distinct phases of the risk assessment process, as explained in the previous chapters.
To facilitate this process, an online Interactive Assessment Tool (IAT) has been developed. The IAT
addresses the first two of these phases; evaluation of novelty, and analysis of risk. This generates
individual risk scores for each risk identified, plus an overall risk score for the TCTP as a whole. The
output from the analysis of risk is used to inform the third phase of the process, to determine
whether or not the TCTP can be made generally available for clinical application on request, or if
further pre-clinical and/or clinical evaluations are required.

3.1. Accessing the IAT


The IAT is accessible on-line (here), and can works in all browsers as long as the equipment has
internet connection.
To be continued

3.2. Key principles for effective use of the EuroGTP II methodologies and IAT
The accuracy and value of the outputs from the IAT will be determined by the accuracy,
comprehensiveness and relevance of the information that is put into it. It is therefore advised that:
i) The process should be treated as a long term exercise; it is unlikely that a single
individual will have sufficient knowledge and expertise to complete the whole process at
one go with no support.
ii) The process should be performed by a team selected to provide the requisite knowledge
and experience to fully identify and evaluate all potential risks. This may include:
 Operational staff
 Scientists developing TCTPs
 Clinicians involved in TCTP provision
 Clinicians making use of TCTPs
This list is not exhaustive.
iii) The initial process can be performed at an early stage in the development of new or
revised TCTPs; this may identify areas of high risk that could be addressed by pre-clinical
development work. The exercise can be repeated in the several different stages of the
implementation of TCTP, in order to re-evaluate the risks based on the information
recollected (by the studies performed and/or relevant references)
iv) There must be a clear understanding of the critical parameters of the TCTP which will
contribute to its safety and efficacy, to enable the risk assessment to be performed
accurately
v) Definition of adequate efficacy /process indicators;
vi) Identification of particular category of the TCTP under analisys;
vii) others TBC
viii)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


20
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

3.3. When should the IAT be used? Commented [a7]: KT comment:

The IAT may be used at any point in the process/product development cycle. Much of the potential Should we merge this two sections (key principles + when should
we use the tool)?
risk inherent in a new product or process can generally be eliminated or ameliorated by well-
Commented [a8]: I am missing something saying that the
planned and focussed pre-clinical studies. It can therefore be useful to use the IAT at a very early funcionalities and methodologies described can also be performed
stage, where it can pinpoint areas where there is a high level of risk that could be addressed with manually using the templates (annexes of the guide)

pre-clinical in vitro studies, or sourcing the appropriate literature. Often at this stage, potential risk As it is described, seems that we cannot use the methodologies if
we don’t use the tool
must be assessed as high due to lack of data. The IAT can be re-run during the development cycle to
evaluate how ongoing work is contributing to ameliorating the overall risk, and identify areas where
further effort should be focussed. If used in this manner, the final use of the IAT prior to proving
products for clinical use will identify the residual risk that can only be addressed with clinical
evaluation or follow up.
The intention is that the IAT will provide the framework for a detailed assessment of risk. It is not
envisioned that the process will be performed quickly, by a single person. Ideally, the assessment
should be performed by a group of individuals selected for their knowledge and experience who will
consider all available information to generate an accurate assessment of risk. It is important that the
rationale for these decisions is recorded and documented.
Note also that the IAT should only be used to assess new risks resulting from the novelty. It is
assumed that for existing TCTPs, which are being provided for clinical use, the existing risks have
been evaluated and are adequately controlled.

Specific guidance applicable to the use of EuroGTP II methodologies and tools for different TCTPs, is
described in the chapters: 4 – Tissues, 5 – HSC, and 6 – ART)

3.4. Step 1 : Evaluation of novelty


The first stage of the tool is the assessment of novelty. This involves answering a series of seven
questions, shown in Table 3.1 below, covering all aspects of the TCTP supply chain from donor
selection to implantation or transfusion of the final product. This stage is intended to generate a
simple ‘yes’ or ‘no’ answer; there is either novelty or not, irrespective of the degree of novelty. If no
novelty is identified, it can be concluded that there is no significant change or innovation in the TCTP
being assessed; in this case, there is no need to proceed with the rest of the IAT.
Specific examples and explanations regarding the interpretation of these questions, are provided in
the specific chapters (Chapter 4 - Tissues, Chapter 5 – HSC, and Chapter 6 - ART).

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


21
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 3.1: Evaluation of novelty (Step 1) Yes No NA

A. Has this type of TCP previously been prepared and issued for clinical use by
your establishment?

B. Will the starting material used to prepare this TCP be obtained from the
same donor population previously used by your establishment for this type
of TCP?

C. Will the starting material for this TCP be procured using a procedure used
previously by your establishment for this type of TCP?

D. Will this TCP be prepared by a procedure (processing, decontamination and


preservation) used previously in your establishment for this type of TCP?

E. Will this TCP be packaged and stored using a protocol and materials used
previously in your establishment for this type of TCP?

F. Will this type of TCP provided by your establishment be applied clinically


using an implantation method used previously?

G. Has your establishment provided this type of TCP for transplantation into
the intended anatomical site before?

3.5. Step 2: Level risk analysis - Identification and Quantification of Risk


If, after completing the step 1, you determine that there is some novelty resulting from your
proposed change, you then proceed to step 2 to identify and quantify the potential risks resulting
from this novelty. There are a number of stages in this process:

Step 2A:Indentification of risk factors


identify the potential risk factors that are relevant to the change. The global risks that should be
considered during this assessment are listed in Table 3.2 below. Specific risk factors, examples and
explanations regarding the interpretation of these risk factors, are provided in the next chapters
(Chapter 4 - Tissues, Chapter 5 – HSC, and Chapter 6 - ART).

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


22
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 3.2 – Identification of Risk Factors

Process Specific risk factors Guidance notes


Consider whether the donor population you intend to obtain the TCTP
from could impart any risk, for example if the TCTP is sourced from an
Donation Donor Characteristics allogeneic donor, there may be risks that immunogenicity could impact
on the clinical performance of the TCTP, and risks of disease
transmission’
‘Consider where and how the TCTP is collected, procured or recovered,
and if this process could have an influence on the TCP. How long does
Recovery process and the process take, how complex is it, and what is quality of the
Procurement
environment’ environment - for example, these factors may impact on the
probability that the TCTP becomes contaminated, or damaged during
recovery
Consider where and how the TCTP is prepared. How long does the
preparation process take and how complex is it – this may impact on
Preparation process and
the risk of contamination, or that it may not be prepared to consistent
environment’
specifications and quality. Also consider the quality of the preparation
environment, which may also affect the risk of contamination
Consider any reagents used during preparation, decontamination,
preservation, storage and transport of the TCTP. Could they damage
Reagents
the TCTP in any way, or could residual traces of reagent remain in the
TCTP that could cause toxic or immunogenic effects in recipients?
Processing/
Consider the risk that the nature of the TCTP, the testing methodology
storing
Reliability of Microbiology and/or the presence of residual processing reagents such as antibiotics
/transport
Testing’ in the finished TCTP may impact the accuracy of any microbiology tests.
Note, this refers specifically to bacteriology/mycology testing of the
TCTP, not any blood tests performed on the donor.
Consider any potential risks arising from how the starting material and
Storage conditions TCTP are stored, between procurement and processing, during
processing, and between processing and clinical application.
Consider any potential risks arising from how the starting material and
Transport conditions TCTP are transported, for example between the sites procurement and
processing, and between the sites of storage and clinical application.
This risk must be considered from the perspective that for some TCTPs,
the presence of intact vital cells is desirable, although it may also
Presence of unwanted
increase risks of, for example, immunogenicity or disease transmission
cellular material
Product This presence might affect to tumour formation, immunogenicity and
disease transmission risks.
Vascular tissues may be more at risk to infiltration by pathogens or
Graft vascularity
malignant cells than avascular tissues
Consider how complex the method of clinical application will be for this
TCTP. How long will it take, and could this introduce risks? What is the
Clinical Complexity of the
scope for errors to be made, and what could the consequences of
Application preparation/application
these errors be?
procedures method
Low feasibility of application standardization might have influence in
the risks of implant failure and disease transmission at least.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


23
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Step 2B:Identification of risks


Consider the potential risks for the risk factors identified above. The potential risks associated with
the clinical use of TCTP comprise:
 Unexpected immunogenicity
 Implant failure / pregnancy loss
 Disease transmission
 Toxicity / Carcinogenicity
 Other potential risks (can be associated with specific TCTP)

Examples of the combination of risk factors and specific risks that may need to be considered are
provided in the TCTP type specific chapters (4, 5 and 6). The purpose of the exercise is to
systematically consider each risk factor and risk in turn against the nature of the change. Note that
for certain combinations of risk factor and specific risk, there may be no relevant examples. It is
recognised that the IAT cannot anticipate all potential types of risk; the specific risk categories listed
for each TCTP type are those which it is generally agreed will be most commonly related to that type.
For any risks not covered by these risk categories, an open, ‘other’ category is provided.

Step 2C: Quantification of Risk


The next step is to perform the risk assessment by determining the Probability, Severity and
Detectability for each risk factor identified for each risk category. Note that:
 There may be more than one risk associated with each risk factor and specific risk. If this is
the case, a single risk assessment must be done to cover multiple risks.
 A explanation of the rationale behind the performed analysis can be detailed and included in
the exercise. This should allow the user to keep record of the risks and risk factors
evaluated.

Assessment of Probability
This assessment requires estimating the probability of any risk occurring. There are five levels of
probability

Table 3.3 – Probability levels1


Level of probability Definition
1 - Rare Difficult to believe it could happen
2 - Unlikely Not expected to happen but possible
3 - Possible May occur occasionally
4 - Likely Probable but not persistent
5 - Almost certain Likely to occur on many occasions

1
Definitions from V&S SoHO Project, 2009 [10]

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


24
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Assessment of Severity
This assessment requires that you estimate the severity of the consequences of the risk, should it
occur. There are four levels of severity.
Table 3.4 – Severity levels2
Level of severity Definition
Mild clinical or psychological consequences for the recipient, however
Non-serious
with no hospitalisation, or anticipated long term consequences/disability
Hospitalisation and/or:
 Persistent/significant disability or incapacity
 Intervention to preclude permanent damage
Serious  Evidence of a serious transmitted infection
 Significant decrease of expected treatment success
 Birth of a child with a infectious or genetic disease following ART
with donor gametes or embryos
 Major intervention necessary to prevent death
 Evidence of a life threatening transmissible infection
Life-threatening
 Birth of a child with life threatening genetic disease following
ART with donor gametes of embryos
Fatal Death of the patient

Assessment of Detectability
This assessment requires that you estimate the probability that the risk will be detected before the
TCTP is applied to and causes harm to recipients.

Table 3.5 – Detectability levels Commented [a9]: Kelly proposes a change in the levels of
detectability
Level of detectability Definition
If we accept this changes we need to correct all chapters and tool
1 - Almost certain (Very The potential defect/harm will almost certainly be detected before
high) clinical application in the recipient
Please give feedback
There is a reasonable chance that the potential defect/harm will be
2 - Moderately high
detected before clinical application in the recipient
There is a low chance that the potential defect/harm will be detected
3- Low
before clinical application in the recipient
It is unlikely that the potential defect/harm will be detected before
4 – Remote (Very low)
clinical application in the recipient
5 - Absolute
The potential defect/harm will be detected only after clinical
uncertainty/Cannot be
application in the recipient
detected

2
Definitions from V&S SoHO Project, 2009 [10]

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


25
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Step 2D: Assessment of Risk Reduction


Based on available evidence, some risks can be reduced. To perform this step, users should choose
the appropriate risk reduction value for each risk.
This assessment requires that you consider any extant data that may support reducing the overall
risk score. This data may support reducing the probability the risk will occur, its severity and/or the
probability that the consequences will not be detected pre-application.
Data that should be taken into account when calculating risk reduction may include:
 Published data in peer reviewed literature
 Internal validation reports
 Clinical outcomes already known

It is also important that the quality and reliability of the data be considered; for example a large scale
clinical trial in a high impact, peer reviewed journal would be considered of high quality and
reliability, whereas unpublished clinical data with limited follow up in a small number of patients less
so.
The GRADE system (https://gdt.gradepro.org/app/handbook/handbook.html) [25] (or similar) can be
used to give an objective evaluation of the quality and reliability of scientific data. Commented [a10]: Kelly Tilleman comment:
Although this step might include some subjectivity, the evidences used to justify the risk reduction If there are randomised controlled trials in the literature available,
should be accurately described in the rationale of the assessment. It is advisable to keep the than the GRADE system
(https://gdt.gradepro.org/app/handbook/handbook.html) [15] (or
references/documents associated with the risk assessment report, in order to easily evidence the similar) can be used to give an objective evaluation of the quality
rationale behind each risk assessment. and reliability of scientific data. However, GRADE is not very
applicable to small studies, single studies, observational data, case
Based on the assessment of the data, different levels of risk reduction can be applied. This is reports, .. which is probably the data that can be found if novelties
with higher risk assessment are addressed. Other quality
accomplished by a percentage reduction in the overall risk (probability x severity x detectability) assessment checklists or bias assessment tools can be used to
calculated in the first three steps of the risk assessment. These levels are shown in Table 3.6 below: consider the quality of the data in these studies.

Can someone confirm?


Table 3.6. – Percentage risk reduction definitions
Percentage risk
Definition
reduction
0 None There is no relevant data available to support reducing the calculated risk score
There is a moderate relevant data available to support reducing the calculated
25 Limited
risk score, based predominantly on unpublished, internal data
There is good quality relevant data available support reducing the calculated
risk score, including published and unpublished data from internal and external
50 Moderate
sources, and some data which has been through an independent peer review
process
There is high quality relevant data to support reducing the calculated risk
75 Extensive
score, including data that has been peer reviewed and published.
There is a substantial amount of high quality relevant data, including multiple
95 Substantial peer reviewed publications, that demonstrates that the probability of the risk
occurring, having a significant impact, and/or being undetected is negligible.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


26
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

On completion of this step, an overall risk score is calculated, which will determine if the risk is
negligible, low, moderate or high.

The level of residual risk will inform whether or not (and what level of) pre-clinical (in vitro & in vivo)
evaluation is indicated for the TCTP, and what level of clinical evaluation and/or follow up will be
proportional to the level of risk estimated.

3.6. Step 3: Definition of extent of studies needed based on the risks quantified
The output from step 2 (A:Indentification of risks factors, B: Identification of risks; C: Quantification
of risks, D: Assessment of risk reduction) will result in the identification and quantification of one or
more residual risks; these can be expressed in the standard format:

‘There is a risk that the TCTP will ………….. due to ………….. resulting in…………..’

E.g. – There is a risk that the TCTP will be immunogenic due to the inadequate removal of donor cells
resulting in an unwanted localised and systemic immune response

Or: - There is a risk that the TCTP will fail due to biomechanical damage caused by the processing
protocol resulting in sudden mechanical failure

The purpose of step 3 is to provide users with guidance as to how to evaluate these risks through the
sequential application of pre-clinical (in vitro, in vivo) and clinical assessments:

Process validation
Process validation is a mandatory activity under the EUTCD, to ensure that a process is reliably
achieving its stated objective. Guidance for performing process validation can be found in the Guide
to the Quality and Safety of Tissues and Cells for Human Application [12], and is not within the scope
of this document. If the final, overall risk is determined to be negligible, no further risk mitigation is
necessary, however, it may be advisable to conduct a revalidation of the process. If the final overall
risk is determined to be low, it is necessary that as a minimum the process is revalidated. However, if
the nature of the risk is not related to the process itself, the requirement for revalidation may not
apply, for example where the novelty is in the method of clinical application.

In vitro Studies
Generally, in vitro assessments will be performed prior to other pre-clinical (in vivo) studies. This
category may also incorporate routine process validation studies. Where the overall risk is low, it is
likely that it can be mitigated purely with in vitro assessments.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


27
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Pre- Clinical Studies

In vivo assessments will usually only be considered where the risk cannot be sufficiently
mitigated with in vitro studies, for cost and ethical reasons. There may however be criteria
that can only be accurately evaluated with in vivo models.

Clinical Studies

If the risk cannot be mitigated to ‘negligible’ or ‘low’ levels by in vitro or pre-clinical studies,
and when ethically accepted, clinical evaluation may be necessary before the TCTP is made
generally available.

Guidance for the correct definition of studies to address the specific risk categories referred in the
step 2, is presented in chapters 4-6.of this document.
The methodology proposed by EuroGTP II, suggests this be done by reference to matrixes and tables.
The matrixes suggest a number of different test criteria, which are again specific for different types
of TCTP, each of which are also subdivided into specific tests. It then suggests which of these tests
could be applied to address specific risk consequences.
The studies proposed in the specific chapters do not intended to be exhaustive, and should be
considered in conjunction with any tests already performed by the TE.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


28
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

4 . Tissues specific chapter

Define which type of TCTP you are evaluating


At first it is important to define for which TCP you are going to use the tool, as this will generate
specific risk factors. In case of Tissues, choose ‘Tissues’ and subsequently which type of tissue is the
subject of the process under evaluation.

FIG 4.1: Diagram of IAT: different types of tissues

If selecting Tissues, you will also be asked to choose a specific anatomical type of Tissue:

 Musculoskeletal
 Cardiovascular
 Amniotic Membrane
 Ocular tissue
 Skin
 Other

4.1. Evaluation of Novelty (Step 1)


This chapter presents the questions as asked when the tool is being used, a brief explanation of what
the question is intended to elicit, and some examples to demonstrate when novelty may or may not
be present, are shown in Table 4.1. below.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


29
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 4.1: Exercise for assessing novelty

Yes No NA
A. Has this type of TCP previously been prepared and issued for clinical
use by your establishment?
Explanation:
The purpose of this question is to determine if your establishment has previously banked or provided the
specific, anatomical type of TCTP for clinical application. It does not require that the TCTP has been banked
using the same process.

Examples:
A1 – Your establishment already banks pulmonary and aortic heart valves, but you intend to start processing
them in a different way. In this case, you would answer ‘Yes’ to this question, and there is no novelty.
A2 – Your establishment already banks Achilles tendon, and you intend to start banking peroneus longus
tendons. In this case you would answer ‘No’; although you already bank tendons, you do not bank this
particular anatomical type of tendon, so there is novelty.
A3 – Your establishment provides pericardial graft as a dural patch, and you intend to start baking fascia lata
for the same purpose. In this case, you would answer ‘No’; although the graft is to be used for the same
purpose for which you already provide another type of graft, you have not banked this type of tissue
previously, so there is novelty.
B. Will the starting material used to prepare this TCP be obtained from
the same donor population previously used by your establishment for
this type of TCP?
Explanation:
This question aims to elicit if there may be differences in the TCTP resultant from the donor population.
Examples of changes that would create novelty are changing the age limits for donors of the TCTP, or
changing specific aspects of the donor selection criteria applicable to the TCTP. Note that this does not
apply to generic changes to donor selection criteria; for example if screening requirements for blood borne
infections are amended, rather it should be considered when making specific changes to donor selection
criteria that impact on specific TCTPs

Examples
B1 – Your establishment wishes to raise the age limit for donors of tendons from 65 to 70. In this case, you
are clearly changing your donor population, so you would answer ‘No’; there is novelty.
B2 – Your establishment implements routine screening of your donor population for a new tropical virus
that has become endemic in your country. In this case, it is a systematic change which will affect donors of
all tissues; whilst you may technically be impacting on your donor population by implementing a new test,
you are not changing the overall makeup of the donor population. You would therefore answer ‘Yes’ to this
question, and there is no novelty.
B3 – Your establishment banks heart valves, and you implement a change in your donor selection criteria
that permits you to accept donors with cardiomyopathy with a non-infectious underlying cause that you had
previously deferred. This is a specific change directed at a specific tissue, which will result in change to your
donor population. You would answer ‘No’ to this question; there is novelty
C. Will the starting material for this TCP be procured using a procedure
used previously by your establishment for this type of TCP?
Explanation:
The question is to determine is to determine if a change in the way in which the TCTP is procured from the
donor (or patient) may impact on its safety or quality

Examples
C1 – Your establishment currently banks skin allografts, which are procured from donors using an electric
dermatome. In order to improve the quality of your grafts, you are proposing to change to a different type of
dermatome. In this case, there may be novelty; you would need to take view, based on your knowledge of the
process, as to whether or not this could introduce significant change
C2 – Your establishments currently retrieves hearts for valve donation from deceased donors within 48 hours
of death; you are considering expanding this time limit to 72 hours. In this case, there is definite novelty, as
there would clearly be risks relating to contamination of the tissue and deterioration of the tissue quality
resulting from the increased post-mortem retrieval time.
D. Will this TCP be prepared by a procedure (processing,
decontamination and preservation) used previously in your

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


30
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

establishment for this type of TCP?


Explanation:
This question covers a wide range of protocols, essentially covering all processes applied to the graft between
retrieval and preservation

Examples:
D1 – Your establishment currently banks tendon allografts which are terminally sterilised with gamma
irradiation; you are considering changing to gas plasma sterilisation. There would clearly be novelty here, as
you are introducing a novel process which could have significant implications for graft safety and quality.
D2 – Your establishment currently used buffered saline in many of your routine tissue processing protocols.
Your current supplier has discontinued this product, and you intend to switch to a new supplier who provides
the reagent to the same specification. In this case, there is unlikely to be novelty; you are not proposing to
make a change to the fundamental process, just changing ‘like with like’.
E. Will this TCP be packaged and stored using a protocol and materials
used previously in your establishment for this type of TCP?
Explanation:
This question seeks to elicit whether there are any significant changes in how the TCTP is packaged and
stored, and distributed prior to transplantation.

Examples:
E1 – Your establishment currently stored bone allografts prior to distribution at -40oC; you are considering
changing this to -20oC. In this case, there is novelty as you are making a change that could clearly affect the
safety and quality of your grafts.
E2 – Your establishment currently provides morsellised bone allografts in 20g pack sizes. You are considering
changing this pack size to 40g. In this case, there is unlikely to be novelty; the change must be one that could
significantly affect the quality and/or safety of the graft.
F. Will this type of TCP provided by your establishment be applied
clinically using an implantation method used previously?
To be completed by the WP6 Leaders

G. Has your establishment provided this type of TCP for transplantation


into the intended anatomical site before?
To be completed by the WP6 Leaders

4.2. Level risk analysis (Step 2)

Step 2A: Identification of risk factors

If, after completing part 1 of the IAT, you determine that there is some novelty resulting from your
proposed change, you should now proceed to step 2 to identify and quantify the potential risks
resulting from this novelty. The risks have been subdivided into 10 factors:

I) Donor Characteristics.
II) Procurement process and environment.
III) Preparation process and environment.
IV) Reagents.
V) Reliability of Microbiology Testing.
VI) Storage Conditions
VII) Transport Conditions.
VIII) Presence of unwanted cellular material
Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015
31
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

IX) Graft vascularity.


X) Complexity of the preparation/application method.

You must first determine which of these risk factors are relevant to the aspect or aspects of your
proposed change which result in novelty. Worked examples are provided later in this document to
demonstrate how the process works.

Step 2B: Identification of risks

Having identified the appropriate risk factor(s), you should then determine which specific risks are
applicable. A standard set of risks is applied to each factor, with an open, ‘other’ category for any
risks not covered in the four main categories.

a) Unwanted immunogenicity
b) Implant failure
c) Disease transmission
d) Toxicity/Carcinogenicity
e) Other

Examples of the combination of risk factors and specific risks that may need to be considered are
provided in the table 4.2. The purpose of the exercise is to systematically consider each risk factor
and risk in turn against the nature of the change. Note that for certain combinations of risk factor
and specific risk, there may be no relevant examples. It is recognised that the IAT cannot anticipate
all potential types of risk; the four specific risks listed are those which it is generally agreed will be
most commonly related to TCTPs. For any risks not covered by these four categories, an open, ‘other’
category may be consider, and is provided in the IAT.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


32
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 4.2. Identification and interpretation of the risk factors and risk associated with tissues

Risks Examples and Explanation Risk Factors Examples and Explanation

Unwanted
TBC WP Leader – Optional/
immunogenicity
• If you increase the age of your donor population, this
could impact on the quality of your graft
This factor requires that Implant failure • Certain aspects of a donor’s medical history may
impact on the suitability of certain grafts for transplantation;
Donation

you consider whether the


Donor Characteristics donor population you changes should be considered in this light.
intend to obtain the TCTP • If a change is made so that a graft that was previously
from could impart any risk only obtained from heart beating donors may not be obtained
Disease transmission
from deceased donors, this may affect the risk of graft
contamination and disease transmission
Toxicity/Carcinogenicity TBC WP Leader
Other TBC WP Leader – Optional/
 Could changes to the procurement process result in
Unwanted elevated quantities of immunogenic material being
This factor requires that immunogenicity present in the graft?
you consider where and
how the TCTP, or the
 Could changes to the procurement process result in
material used to Implant failure
Procurement

the grafts being damaged during procurement?


manufacture it, are
Procurement process and  Could changes to the procurement process result in
recovered. For example,
environment an increased risk of donor-recipient disease
how long does the process
transmission?
take, how complex is it, Disease transmission
and what is quality of the  Could changes to the procurement process result in
an increased risk of the graft being contaminated with
environment?
environmental organisms?
 Could any chemicals (e.g. disinfectants) used in the
Toxicity/Carcinogenicity
procurement process be transferred to the graft?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


33
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Other TBC WP Leader – Optional/
Unwanted
TBC WP Leader
immunogenicity
 Could the length of the process result in the quality of
This factor requires that the graft deteriorating?
you consider where and Implant failure  Could the environmental conditions applied during
how the TCTP is prepared. the preparation process (e.g. heat, pressure, humidity
Preparation process and For example, how long etc. affect the graft quality?
environment and how complex is
 Could the length, complexity or environment where
preparation process, and
Disease transmission the preparation process takes place affect the risk of
what is the quality of the
environmental contamination?
preparation environment?
 Could the TCTP degrade during the preparation,
Toxicity/Carcinogenicity
generating toxic compounds?
Processing/ storing /transport

Other TBC WP Leader – Optional/


This factor requires that Unwanted
TBC WP Leader
you consider any reagents immunogenicity
used during recovery, Implant failure TBC WP Leader
preparation, Disease transmission TBC WP Leader
decontamination and
Toxicity/Carcinogenicity TBC WP Leader
storage of the TCTP. For
example, cCould they
Reagents
damage the TCTP in any
way, or could residual
traces of reagent remain Other TBC WP Leader – Optional/
in the TCTP that could
cause toxic or
immunogenic effects in
recipients?
This factor requires that Unwanted
TBC WP Leader
Reliability of Microbiology you consider the risk that immunogenicity
Testing. the nature of the TCTP, Implant failure TBC WP Leader
the testing methodology Disease transmission TBC WP Leader
and/or the presence of Toxicity/Carcinogenicity TBC WP Leader

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


34
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
residual processing
reagents such as
antibiotics in the finished
TCTP may impact the
accuracy of any
microbiology tests. Note, Other TBC WP Leader – Optional/
this refers specifically to
bacteriology/mycology
testing of the TCTP, not
any blood tests performed
on the donor.
This factor requires that Unwanted
TBC WP Leader
you consider any potential immunogenicity
risks arising from how the Implant failure TBC WP Leader
starting material and TCTP Disease transmission TBC WP Leader
are stored, between
Storage Conditions Toxicity/Carcinogenicity TBC WP Leader
procurement and
processing, during
processing, and between Other TBC WP Leader – Optional/
processing and
implantation.
This factor requires that Unwanted
TBC WP Leader
you consider any potential immunogenicity
risks arising from how the Implant failure TBC WP Leader
starting material and TCTP Disease transmission TBC WP Leader
are transported, for
Transport Conditions Toxicity/Carcinogenicity TBC WP Leader
example between the sites
of procurement and
processing, and between Other TBC WP Leader – Optional/
the sites of storage and
implantation.
Presence of unwanted This factor requires that Unwanted
Produ

TBC WP Leader
cellular material. you consider the risk that immunogenicity
ct

for some TCTPs, the Implant failure TBC WP Leader

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


35
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
presence of intact vital Disease transmission TBC WP Leader
cells is desirable, although Toxicity/Carcinogenicity TBC WP Leader
it may also increase risks
of, for example,
immunogenicity or disease Other TBC WP Leader – Optional/
transmission.

This factor requires that Unwanted


TBC WP Leader
you consider the risk that immunogenicity
vascular tissues may be Implant failure TBC WP Leader
Graft vascularity more at risk to infiltration Disease transmission TBC WP Leader
by pathogens or malignant
Toxicity/Carcinogenicity TBC WP Leader
cells than avascular
tissues. Other TBC WP Leader – Optional/
This factor requires that Unwanted
Clinical application procedure

TBC WP Leader
you consider how complex immunogenicity
the method of Implant failure TBC WP Leader
implantation will be for Disease transmission TBC WP Leader
Complexity of the this TCTP. How long will it
Toxicity/Carcinogenicity TBC WP Leader
preparation/application take, and could this
method introduce risks? What is
the scope for errors to be
made, and what could the Other TBC WP Leader – Optional/
consequences of these
errors be?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


36
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Step 2C: Quantification of risks

When the risk factors are selected and the potential harm is identified, the potential impact of
this risk needs to be determined:

i. What is the probability that the harm due to the risk factor will occur (based on V&S
SoHO Project [10]):

Table 5.3 .. – Probability levels3


Level of probability Definition
1 - Rare Difficult to believe it could happen
2 - Unlikely Not expected to happen but possible
3 - Possible May occur occasionally
4 - Likely Probable but not persistent
5 - Almost certain Likely to occur on many occasions

ii. What is the severity of the outcome, should the harm occur

Table 5.4. – Severity levels4


Level of severity Definition
Mild clinical or psychological consequences for the recipient, however
Non-serious
with no hospitalisation, or anticipated long term consequences/disability
Hospitalisation and/or:
 Persistent/significant disability or incapacity
Serious  Intervention to preclude permanent damage
 Evidence of a serious transmitted infection
 Significant decrease of expected treatment success
 Major intervention necessary to prevent death
Life-threatening
 Evidence of a life threatening transmissible infection
Fatal Death of the patient

iii. What is the detectability of identifying the risk The detectability is “the chance for
the potential effect to be detected”. It refers to the probability of detection of the
potential effect before producing any harm to the recipients.

Table 5.5. – Detectability levels


Level of detectability Definition
The potential defect/harm will almost certainly be detected before
1 - Almost certain
clinical application in the recipient (for example: poor cell viability)
There is a reasonable chance that the potential defect/harm will be
2 - Moderately high
detected before clinical application in the recipient

3
Definitions from V&S SoHO Project, 2009 [10]
4
Definitions from V&S SoHO Project, 2009 [10]

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


37
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

There is a low chance that the potential defect/harm will be


detected before clinical application in the recipient (potential
3- Low
defect/harm requires additional quality controls and may not be
detected by the current available techniques)
It is unlikely that the potential defect/harm will be detected before
clinical application in the recipient (for example, it could be that
4 - Remote
there are epigenetic changes in the cell product that cannot be
determined).
The potential defect/harm will be detected only after clinical
5 - Absolute uncertainty application in the recipient (for example, an increased possibility of
an severe immunogenetic reaction).

Step 2D: Assessment of risk reduction

To what degree can this risk be reduced by pre-existing data5

Risk reduction can be take into account when there is data evidence that supports reducing the
overall risk score. This may be (for example) published data in the peer reviewed literature,
internal validation reports, clinical outcomes, or any other relevant sources of data. The quality
of the data should also be taken into consideration, for example data in a high impact factor,
peer reviewed publication should be given more weight than unpublished data.
The risk estimated when implementing novelties may be reduced by:

 Bibliographic references that demonstrate safe and efficacy of products and/or


techniques;
 Technical improvements that may decrease the risk associated with the activities (e.g.:
when the novel process represents a change of processing method from “open system”
to a “close system”, the risk factor “time of exposure” may represent a reduction of
risks when processing with the novel method.)
Based on your evaluation of the totality of the evidence, different levels of risk reduction
may be assign according the scale:

Table 5.6.– Percentage risk reduction definitions


Percentage
risk Definition
reduction
There is no relevant data available to support reducing the calculated risk
0
score
25 There is a moderate relevant data available to support reducing the calculated

5
Pre-existing data is defined as data generated either by your organization or by others that indicates
the probability or severity score may be lower than the score you have selected, or that the likelihood of
detection may be higher than the score you have selected.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


38
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

risk score, based predominantly on unpublished, internal data


There is good quality relevant data available support reducing the calculated
risk score, including published and unpublished data from internal and
50
external sources, and some data which has been through an independent peer
review process
There is high quality relevant data to support reducing the calculated risk
75
score, including data that has been peer reviewed and published.
There is a substantial amount of high quality relevant data, including multiple
95 peer reviewed publications, that demonstrates that the probability of the risk
occurring, having a significant impact, and/or being undetected is negligible.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


39
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

4.3. Definition of extent of studies needed based on the risks quantified (Step 3)

4.3.1 Pre-clinical evaluation (chapters for different types of tissue)


Step 3A: Risk reduction strategies

Testing Matrix: In vitro testing methodologies appropriate to address different risks (This matrix maybe Generic.. still needs to be completed and review by our
partners)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

procurement or
acquired during
Carcinogenicity

Teratogenicity
integrate with

Blood borne
Anaphylaxis

cytotoxicity

cytotoxicity
mechanical

mechanical
host tissue

processing
Infections
infections
Failure to
Test criteria Specific test
Localised

Localised
response

response
Systemic

Systemic
Immune

immune

Gradual

Sudden
failure

failure
Aseptic
fill/handling
Decontaminati
on efficacy
(m.o
logarithmic
reduction)
Osteoinductivi
ty test
Reological test
Process
Biomechanical
qualification
test
Decellularizati
on process
efficacy
Residual
moisture
Morphology
test (p.e. skin
thickness,
ocular OTC)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


40
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Materials and
reagents
quality specs
definition
Packaging
sealing test
Stability test
Validation of
the
microbiology
analytical
method
Validation of
Analytical the
methods osteoinduction
related tests test
Validation of
the cellular
and or DNA
traces
analytical
method
Extract
In vitro cytotoxicity
cytotoxicity Contact
cytotoxicity
Dye exclusion
assays
Donor cell Metabolic
viability activity assays
Live/dead
staining
Expression of
cell surface
Donor cell markers
functionality Quantification
of secreted
factors
Histological Safranin O
evaluation of (proteoglycans
the ECM & GAGs)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


41
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Alizarin Red or
Von Kossa
(Calcium)
Van Gieson
(Collagen)
Periodic
acid/Schiff
(Collagen)
Pico Sirius Red
(Collagen)
Inter fibre
space
Histological
evaluation of H&E stain
cell content
Quantitative
Residual DNA testing
content Qualitative
testing (DAPI)
Collagenase
Biochemical
resistance
evaluation of
Collagen
ECM quality
nativity
Ultimate
tensile stress
Ultimate
tensile strain
Biomechanical Cyclic testing
properties Suture pullout
Pressure
testing
Three point
bending
Competency
Haemodynami
In vitro
cs
functionality
Osteoinductivi
ty
Residual HPLC
processing Mass
reagents spectrometry

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


42
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Biochemical
tests
Immunohisto-
chemistry

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


43
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

In vivo testing methodologies appropriate to address different risks (Draft example)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

procurement or
acquired during
Carcinogenicity

Teratogenicity
integrate with

Blood borne
Anaphylaxis

cytotoxicity

cytotoxicity
mechanical

mechanical
host tissue

processing
Criteria Specific test

Infections
infections
Failure to
Localised

Localised
response

response
Systemic

Systemic
Immune

immune

Gradual

Sudden
failure

failure
Cellular infiltration
(histology)
Biocompatibility
Bone induction
chamber
Encapsulation
Immunological
Blood tests
response
Calcification
Thrombotic Blood test
response Doppler ultrasound
Echocardiogram
Regurgitation
Functionality
Tissue regeneration
Engraftment
In vivo
carcinogenicity
In vivo
teratogenicity
General wellbeing
(alive and well, sick,
dead)
Health
Weight change
Mobility
Infection
histological analysis
Bone microcomputed
regeneration tomography (micro-
CT) analysis

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


44
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

mechanical test
analysis
radiograph analysis
sequential
fluorescence
labelling analysis
dynamic contrast-
enhanced magnetic
resonance imaging
Valves thrombogenicity
functionality fluid dynamics
structural integrity
calcification
Ocular imaging (optical
functionality coherence
tomography (OCT)
In vivo functional
assessment: a)
electroretinography
(pERG), b) visual
evoked potentials
(VEP)
Morphological
assessment
(histology, IHC, EM,
stereology)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


45
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

4.3.2. Clinical evaluation and follow up plans

Step 3B: Definition of Clinical Studies

(chapters for different types of tissue)

1. Bone - Testing Matrix – Clinical Test Summary (Draft example to be completed and reviewed)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

procurement or
acquired during
Carcinogenicity

Teratogenicity
integrate with

Blood borne
Anaphylaxis

cytotoxicity

cytotoxicity
mechanical

mechanical
host tissue

processing
Test category Detailed investigational options

Infections
infections
Failure to
Localised

Localised
response

response
Systemic

Systemic
Immune

immune

Gradual

Sudden
failure

failure
1. Radiography - Heterotopic
ossification
2. Radiography - Localised repair
e.g. callus formation
3. Radiography - Cortical
regeneration
Physical 4. Radiography - Trabecular
investigation resorbtion
(discrete 5. Radiography - Trabecular
outcome incorporation
measures with 6. Radiography - Stem subsidence
quantifiable (hip implant), with the RSA
results) technique
7. Radiography - Loosening
(implant placement)
8. Radiography – bone
mineralisation (DBM)
9. Allosensetisation – Antibody
profile

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


46
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

1. Spinal curve
Overall Clinical
outcome 2. Weight bearing
measures
3. Functional measures

1. QoL indicators e.g. SF-36


Patient
Reported 2. Self assessment tests e.g.
outcome Oxford, WOMAC, AAOS
measures 3. Neck Disability Indicator
(Cervical spinal fusion)
1. Arthroplasty revision rate
Procedure or
2. Dislocation rate
graft failure
3. Re-operation rate

1. Pain

2. Post-op infection
Post operative
complications 3. Hematoma

4. Thromoembolic complications

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


47
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

4.4. Worked examples

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


48
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

5. Hematopoietic Stem Cell Specific Chapter

Define which type of TCTP you are evaluating

At first it is important to define for which TCP you are going to use the tool, as this will
generate specific risk factors. In case of Hematopoietic Stem Cells (HSC), choose which type of
cells is the subject of the process under evaluation.

Figure 5.1: Diagram of IAT: different types of HSC

If selecting HSC, you will also be asked to choose a specific type of Cells under evaluation:

 From Bone Marrow


 From Peripheral Blood
 From Cord Blood
 From other sources

5.1. Evaluation of Novelty (Step 1)


This chapter presents the questions as asked when the tool is being used, a brief explanation
of what the question is intended to elicit, and some examples to demonstrate when novelty
may or may not be present. The questions as they appear in the IAT are shown in Table 5.1
below.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


49
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 5.1: Exercise for assessing novelty

Yes No NA
A. Has this type of TCP previously been prepared and issued for
clinical use by your establishment?
Explanation:
The purpose of this question is to determine whether your institution has previously prepared, and
issued the specific, anatomical type of TCTP in clinical application for a specific indication. It does not Commented [AB11]: Je gebruikt afwisselend TCP en TCTP??
require that the TCTP has been issued and administered before for a different indication. Commented [WAv12]: Zal het rechttrekken.
Examples:
Commented [WAv13]: Zelfde product, maar voor andere
A1 – Your establishment is already performing T-cell depletion on haematopoietic grafts, but you
indicatie. Bv stamcelproduct dat voor hematologische maligniteit
intend to revise the processing. In this case you would answer ‘Yes’ to this question, and there is no wordt ingezet, wordt ingezet voor behandeling metabole ziekte.
novelty.
Commented [AB14]: Wat bedoel je hier mee? Zelfde ndere
A2 - Your establishment is performing haematopoietic stem cell transplantation (HSCT) using bone indicatie
marrow (BM) and peripheral stem cell (PBSC) grafts. It is decided to start a cord blood transplantation
Commented [WAv15]: Zelfde product, maar voor andere
programme. In this case you answer ‘No’; you have no experience in handling and issuing cord blood. indicatie. Bv stamcelproduct dat voor hematologische maligniteit
A3 – wordt ingezet, wordt ingezet voor behandeling metabole ziekte.

B. Will the starting material used to prepare this TCP be obtained


from the same donor population previously used by your
establishment for this type of TCP?
Explanation:
This question aims to elicit possible differences in the characteristics of the TCTP caused by a change
in the donor population. Examples of changes that would create novelty are changing the age limits
for donors of the TCTP, or changing specific aspects of the donor selection criteria applicable to the
TCTP. Note that this does not apply to generic changes to donor selection criteria; for example if
screening requirements for blood borne infections are amended. It , rather should be considered
when making specific changes to donor selection criteria that has an impact on specifications of the
TCTP’s.
Examples:
B1 – Your establishment wishes to raise the age limit for donors of haematopoietic stem cells from 70
to 75. In this case, you are clearly changing your donor population, so you would answer ‘No’; there is
a novelty.
B2 – Your establishment implements routine screening of your donor population for a new virus that
has become endemic in your country. In this case, it is a systematic change which will affect donors of
all tissues; whilst you may technically may have an impact on your donor population by implementing
a new test, you are not changing the overall makeup of the donor population. You would therefore
answer ‘Yes’ to this question, and there is no novelty.
B3 – Your organisation decides to start immunization of donors prior to stem cell donation. This is a
specific change directed at the immune system of donor and recipient, which will result in change to
your donor population characteristics . You would answer ‘No’ to this question; there is a novelty.
C. Will the starting material for this TCP be procured using a
procedure used previously by your establishment for this type of
TCP?
Explanation:
The question is to determine whether a change in the way in which the TCTP is procured from the
donor (or patient) may impact on its safety or quality.
Examples:
C1 – Your establishment is currently administering filgrastim (G-CSF) for the mobilization of
haematopoietic stem cells in donors. It is decided to start using a biosimilar for this purpose. In this
case, there may be a novelty; because the nature of the cells and composition of the graft could have
been changed in a way that it influences the quality and efficacy .
C2 : Your establishment decides to change the apheresis kits/system from brand A to brand B. Both
devices have CE marking for collection for stem cells and are used in other establishments . The
collection technique is based on the same principles. This is not a novelty , because the procedure has

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


50
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

shown to be suitable for the purpose and the technique is not new in your hands
D. Will this TCP be prepared by a procedure (processing,
decontamination and preservation) used previously in your
establishment for this type of TCP?
Explanation:
This question covers a wide range of protocols, essentially covering all processes applied to the graft
between retrieval and preservation.
Examples:
D1 – Your establishment currently storing autologous PBSC grafts in liquid nitrogen storage, after
controlled-rate freezing. You are considering changing to mechanical freezing and storage. There
would clearly be novelty here, as you are introducing a novel process which could have significant
implications for graft safety and quality.
D2 – Your establishment currently used buffered saline in many of your routine cell processing
protocols. Your current supplier has discontinued this product, and you intend to switch to a new
supplier who proved the reagent to the same specification. In this case, there is unlikely to be a
novelty; you are not proposing to make a change to the fundamental process, just changing ‘like with
like’.
E. Will this TCP be packaged and stored using a protocol and
materials used previously in your establishment for this type of
TCP?
Explanation:
This question seeks to elicit whether there are any significant changes in how the TCTP is packaged
and stored, and distributed prior to transplantation.
Examples:
E1 – Your establishment currently transports BM grafts by room temperature.. You are considering to
change the procedure and transport all HPC BM and A products cooled (4-10°C). There would clearly
be a novelty, as you are making a change that could affect the safety and quality of your grafts.
E2 – Your establishment is adding a tempex box to protect the stem cell bag during transport. There is
no novelty because the box does not influence the essential characteristics of the product.
F. Will this type of TCP provided by your establishment be applied
clinically using an implantation method used previously? Commented [AMvW16]: Find this one difficult: a ‘new’
Explanation: indication (without evidence for efficacy) is NOT a novelty?
This question seeks to elicit whether there are any significant changes in how the TCTP is clinically
applied.
Examples:
F1 – Your establishment currently administered cord blood stem cells intravenously. It is considered
to start direct intra-bone infusion. In this case there is a novelty, the safety and efficacy of the
changed method has to be proved.
G. Has your establishment provided this type of TCP for
transplantation into the intended anatomical site before?
Explanation:
This question seeks to elicit whether there are any significant changes in how the TCTP is clinically
applied.
Examples:
G1 – Your establishment currently provides the TCTP for haematological malignancies via
intravenous infusion . It is considered to start a programme for the use of this TCTP for cardiovascular
repair by direct infusion into affected areas of the heart muscles In this case you answer ‘Yes’, there is
no novelty
.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


51
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

5.2. Level risk analysis (Step 2)


The 2nd exercise aims to determine the risk associated with the novelties attenuated in the
process being evaluated.

Every modification in the processes associated with the donation, procurement, testing,
processing, storage, and distribution of TCTP may have potential consequences for the quality
of these products and safety of recipients. Moreover, different levels of novelties represent
different risks and distinct impact in the quality and safety of the tissue and cell products. The
evaluation of the different levels of these risks can be performed using the methodology
proposed in the current rationale.

Step 2A: Identification of risk factors

At first , the risk factors associated with the changes in the process are selected . There are
nine risk factors that could apply to HPC when hematopoietic cells are concerned (table 5.2).

You must first determine which of these risk factors are relevant to the aspect or aspects of
your proposed change which result in novelty. Worked examples are provided later in this
document to demonstrate how the process works.

Step 2B: Identification of risks

Having identified the appropriate risk factor(s), you should then determine which specific risks
are applicable. A standard set of risks is applied to each factor, with an open, ‘other’ category
for any risks not covered in the four main categories.

a) Unwanted immunogenicity
b) Engraftment failure
c) Disease transmission
d) Toxicity/Carcinogenicity
e) Other

Examples of the combination of risk factors and specific risks that may need to be considered
are provided in the table 5.2. The purpose of the exercise is to systematically consider each risk
factor and risk in turn against the nature of the change. Note that for certain combinations of
risk factor and specific risk, there may be no relevant examples.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


52
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 5.2. Identification of the risk factors and risks associated with HSC

Risks Explanation Risk Factors Examples

 Could adjustment of donor selection criteria


Unwanted immunogenicity (age, HLA match grade), induce (severe) Graft
versus Host Disease?

Consider whether the novelty in  Could increasing the age of the donor population
your process has an impact at the impact the quality of the graft?
moment of the donation. Engraftment failure  Could certain aspects of a donor’s medical
Donation

This factor requires that you history impact the number of HPCs before
Donor Characteristics
consider whether the donor transplantation?
population you intend to obtain the  Is the risk for transmission of infectious diseases
TCTP from, could cause any risk for Disease transmission increased if you accept donors who travelled
the recipient endemic areas
 Could the use of a new type of bag to collect the
Toxicity/Carcinogenicity
graft induce toxicity?
 Will the use of a new apheresis device affect the
Other
number of HPC collected?
Consider where and how the TCT P  Could changes to the procurement process
is recovered currently and whether Unwanted immunogenicity result in elevated quantities of immunogenic
the changes proposed with the material being present in the graft?
Procurement

novel method changes recovery  Could the use of new hematopoietic growth
Procurement process time, complexity, quality of the Engraftment failure factors affect the composition of the graft, and
and environment environment? resulting in poor engraftment?
For example, how long does the  Could changes to the procurement process
process take, how complex is it, and Disease transmission result in an increased risk of donor-recipient
what is how does the procurement disease transmission?
devices affect the quality of the Toxicity/Carcinogenicity  Could any chemicals (e.g. disinfectants) used in

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


53
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
HPC? the procurement process be transferred to the
graft?
 Does a different collection needle influences the
Other
number of specific type of cells? (CD 34??)
Unwanted immunogenicity
 Could the length of the process result in the
Consider the current processing quality of the graft deteriorating?
method for the TCTP how the
 Could the environmental conditions applied
novelty in processing can affect the Engraftment failure
during the preparation process (e.g.
product. How long does the novel
temperature, pressure, humidity) affect the graft
preparation process take and how
quality?
complex is it – this may have an
 Could the length, complexity or environment
Preparation process and impact on the risk of
where the preparation process takes place affect
environment contamination, or cell
the risk of environmental contamination
characteristics that may not be Disease transmission
Processing/ storing /transport

 Could changes to the procurement process


consistent with product
result in an increased risk of the graft being
specifications. Also consider the
contaminated with environmental organisms?
quality of the preparation
environment, which may also affect  Could the TCTP degrade during the preparation,
Toxicity/Carcinogenicity
the risk of contamination. generating toxic compounds?
 Can the devices use in the processing influence
Other
the quality of the HPC ?
 Could change of cryoprotectant induce an
Unwanted immunogenicity
Consider any reagents used during unwanted immunogenic reaction?
recovery, preparation,  Could change of cry protectant affect
Engraftment failure
decontamination, and storage of engraftment?
the TCTP. Could they damage the  Could the use of reagents lead to
Reagents Disease transmission
TCTP in any way, or could residual decontamination of the graft?
traces of reagent remain in the  Could the use of reagents cause toxic effects in
TCTP that could cause toxic or Toxicity/Carcinogenicity
the recipient?
immunogenic effects in recipients.
Other 

Reliability of Consider the risk that the testing


Unwanted immunogenicity
Microbiology Testing. methodology and / or presence of

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


54
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
residual processing reagents such
as antibiotics in the finished TCTP Engraftment failure
may impact the accuracy of any
 Could the change of processing medium mask a
microbiology/mycology testing of
Disease transmission positive outcome of current microbiology
the TCTP. This risk factor is not
testing?
about blood tests on the donor.
Toxicity/Carcinogenicity

Other

 Can a change in the plastics of primary packaging


Unwanted immunogenicity cause enhanced immunogenic material in the
Consider any potential risks arising grafts
from how the starting material and  Could the storage temperature affect the
Engraftment failure
TCTP are stored, between viability of the cells?
Storage Conditions  Could the storage temperature affect the risk of
procurement and processing, Disease transmission
during processing, and between contamination?
processing and implantation.  Can the cryoprotectant cause toxic reactions in
Toxicity/Carcinogenicity
the recipient of the graft?
Other 
Unwanted immunogenicity
 Can the duration of the shipment influence the
Consider any potential risks arising Engraftment failure
number of relevant cells present in the graft ?l
from how the starting material and
 Could the duration of the transport induce the
TCTP are transported, for example Disease transmission
Transport Conditions risk of contamination?
between the sites of procurement
and processing, and between the Toxicity/Carcinogenicity
sites of storage and implantation.  Can shaking and mechanical movements caused
Other by a new transport method hamper the integrity
of the packaging?
Presence of unwanted Consider the risk of s the presence  Do centrifugation forces during apheresis cause
uct
od

Unwanted immunogenicity
Pr

cellular material. of intact ivated cells, debris or cell the presence of cell debris?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


55
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
components which may cause , Engraftment failure
immunogenicity or disease
transmission.  Can the recipient be infected by due to
Disease transmission contamination of the cord blood during
procurement?
Toxicity/Carcinogenicity

Other

Consider how complex the method Unwanted immunogenicity


Clinical application

of implantation will be for this TCTP. Engraftment failure


procedure

Complexity of the How long will it take, and could this


preparation/application introduce risks? What is the scope Disease transmission
method for errors to be made, and what
could the consequences of these Toxicity/Carcinogenicity
errors be? Other

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


56
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Step 2C: Quantification of risks

When the risk factors are selected and the potential harm is identified, the potential impact of
this risk needs to be determined:

iv. What is the probability that the harm due to the risk factor will occur (based on V&S
SoHO Project [10]):

Table 5.3 .. – Probability levels6


Level of probability Definition
1 - Rare Difficult to believe it could happen
2 - Unlikely Not expected to happen but possible
3 - Possible May occur occasionally
4 - Likely Probable but not persistent
5 - Almost certain Likely to occur on many occasions

v. What is the severity of the outcome, should the harm occur

Table 5.4. – Severity levels7


Level of severity Definition
Mild clinical or psychological consequences for the recipient, however
Non-serious
with no hospitalisation, or anticipated long term consequences/disability
Hospitalisation and/or:
 Persistent/significant disability or incapacity
Serious  Intervention to preclude permanent damage
 Evidence of a serious transmitted infection
 Significant decrease of expected treatment success
 Major intervention necessary to prevent death
Life-threatening
 Evidence of a life threatening transmissible infection
Fatal Death of the patient

vi. What is the detectability of identifying the risk The detectability is “the chance for
the potential effect to be detected”. It refers to the probability of detection of the
potential effect before producing any harm to the recipients.

Table 5.5. – Detectability levels


Level of detectability Definition
The potential defect/harm will almost certainly be detected before
1 - Almost certain
clinical application in the recipient (for example: poor cell viability)
There is a reasonable chance that the potential defect/harm will be
2 - Moderately high
detected before clinical application in the recipient

6
Definitions from V&S SoHO Project, 2009 [10]
7
Definitions from V&S SoHO Project, 2009 [10]

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


57
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

There is a low chance that the potential defect/harm will be


detected before clinical application in the recipient (potential
3- Low
defect/harm requires additional quality controls and may not be
detected by the current available techniques)
It is unlikely that the potential defect/harm will be detected before
clinical application in the recipient (for example, it could be that
4 - Remote
there are epigenetic changes in the cell product that cannot be
determined).
The potential defect/harm will be detected only after clinical
5 - Absolute uncertainty application in the recipient (for example, an increased possibility of
an severe immunogenetic reaction).

Step 2D: Assessment of risk reduction

To what degree can this risk be reduced by pre-existing data8

Risk reduction can be take into account when there is data evidence that supports reducing the
overall risk score. This may be (for example) published data in the peer reviewed literature,
internal validation reports, clinical outcomes, or any other relevant sources of data. The quality
of the data should also be taken into consideration, for example data in a high impact factor,
peer reviewed publication should be given more weight than unpublished data.
The risk estimated when implementing novelties may be reduced by:

 Bibliographic references that demonstrate safe and efficacy of products and/or


techniques;
 Technical improvements that may decrease the risk associated with the activities (e.g.:
when the novel process represents a change of processing method from “open system”
to a “close system”, the risk factor “time of exposure” may represent a reduction of
risks when processing with the novel method.)
Based on your evaluation of the totality of the evidence, different levels of risk reduction
may be assign according the scale:

Table 5.6.– Percentage risk reduction definitions


Percentage
risk Definition
reduction
There is no relevant data available to support reducing the calculated risk
0
score
25 There is a moderate relevant data available to support reducing the calculated

8
Pre-existing data is defined as data generated either by your organization or by others that indicates
the probability or severity score may be lower than the score you have selected, or that the likelihood of
detection may be higher than the score you have selected.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


58
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

risk score, based predominantly on unpublished, internal data


There is good quality relevant data available support reducing the calculated
risk score, including published and unpublished data from internal and
50
external sources, and some data which has been through an independent peer
review process
There is high quality relevant data to support reducing the calculated risk
75
score, including data that has been peer reviewed and published.
There is a substantial amount of high quality relevant data, including multiple
95 peer reviewed publications, that demonstrates that the probability of the risk
occurring, having a significant impact, and/or being undetected is negligible.

5.3. Definition of extent of studies needed based on the risks quantified (Step 3)
Using the EuroGTP II methodologies you will be able to perform a risk analysis, determine the
risk profile and the level of risk associated with the novel process or procedure. As a result the
tools (IAT / EuroGTP II algorithm) will provide the total risk value (number) and the final risk
score which is proportional to the number of risk evaluated (in the form of a level of risk).

It is important to state that HPC transplant centres should be prepared to invalidate treatment
when proven problematic (in terms of safety and effectiveness) even when a novelty of
negligible risk was implemented. HPC transplant centres should collect data and register of
follow-up in a systematic way and make them available to the scientific community regardless
of the success of the treatment: not withholding results that point to a negative outcome or
that turn out to be inconclusive. Therefore it is important in all processes, regardless of the level
of risk, to monitor and register SARE / SAE.

The table below provides general guidance on the follow-up studies needed in term of the level
of risk determined (adjusted according to Provoost V. et al. 2014).

Level of Risk Extend of Studies needed


A change in process could have a negligible level of risk because it is part of a
therapy or procedure that is considered as established or standard.

In this case multi-centred studies (ideally RCT) are published in peer-


reviewed journal and the procedures are performed according to a validated
and standard protocol. Minimal process validation is needed. The technical
performance of staff should be monitored and comparable with other TE or
Negligible published studies, therefore standard Key Performance indicators (KPI)
should be monitored on the technical quality of the staff performing the
procedures. Dropping KPIs indicating protocol drift must lead to investigation
of both the procedural steps and / or the possibility to re-train staff.

A routine/safety follow up program is enough as the good practices state.


Follow-up procedures should be focused on assessing efficacy, comparing
the clinical follow-up with the results obtained before the implementation of

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


59
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

the change in the process. Long-term (ideally trans-generational) health


effects, including aspects such as fertility, oncology and mental health should
be monitored.

Implementing a standard procedure or treatment in an ART centre that has


never performed this procedure exerts an intensive validation. Training of
staff is necessary in order to reach the outcomes published in scientific
literature. Having a mentor/mentee relationship with an ART centre having
experience is highly recommended. Specifications on performance should be
determined and when these limits are met by training on spare tissues and
cells, staff can be authorized for performing the procedure.

A learning curve might be expected and should be part of the validation


report. When implementing the procedure, additional quality controls must
be performed to monitor Critical Process Parameters (CPPs) and Critical
Qualtiy Attributes (CQAs). For example, when a TE is switching from T-cell
depletion (TCD) to CD34+_selection (which they never performed before),
engraftment rare, and graft versus host reactions should be carefully
Low monitored in relation to the staff performing the procedure.

A safety follow up program is necessary. Follow-up procedures should be


focused on assessing efficacy, comparing the clinical follow-up with the
results obtained before the implementation of the change in the process and
in relation to the results published in scientific literature. As the procedure or
treatment encompasses an established or standard technique. The expected
learning curve should be kept as short as possible and put in relation to the
follow up program.

Likewise, established techniques are prone to long-term (ideally trans-


generational) follow-up of the health effects. TE or ORHA implementing an
established technique shall perform long-term follow-up and could base
their follow-up items on the mentor facility. This way of working could lead
to periodic evaluation of performance in the mentor/mentee relationship.

Novel procedures or treatments that exert a moderate risk and are


considered innovative. The treatment has shown proof of principle and there
is reassuring data in literature in terms of both safety and effectiveness at
least in animal studies and pre-clinical data shows normal engraftment or
response. The studies that have published this data should have a sound
methodology and published in peer-reviewed journals.

In order to implement an innovative treatment, an enhanced validation is


Moderate
necessary including and a range of additional quality controls performed to
monitor Critical Process Parameters (CPPs), Critical Quality Attributes (CQAs),
and the impact of the implemented TCTP should be carefully monitored.
Since reassuring data of this innovative treatment is already available, a more
specific monitoring of the published critical parameters can be performed
instead of a registration of all critical parameters.

Clinical evaluation and follow up programs should be implemented to assess

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


60
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

reassuring mid-term safety (3 months up to 5 years post-delivery including


data on psychological wellbeing) and these studies should refer to patients
undergoing the procedure as well as the donors where applicable.

A new procedure can be offered to patients in an experimental design aiming


at showing proof of principle, short-term safety and/or effectiveness.

An extensive validation including and a range of additional quality controls


performed to monitor Critical Process Parameters (CPPs), Critical Quality
Attributes (CQAs), and the impact of the implemented changes is required.
This extensive validation should include:

Non clinical studies: preferably there should be studies showing the


experimental procedure is safe in animals.
High
Pre-clinical Studies: when experimental treatments encompass a laboratory
phase, then at least the viability of cells should be looked at in detail,
monitored and registered.

Follow up program: experimental treatments should only be offered to a


selected and limited patient cohort and these patients should be clearly
informed on the experimental status and should receive information about
(the lack of knowledge about) possible risks, alternative treatments etc.
ORHAs should only offer experimental treatments or treatments based on
experimental procedures after approval by a commission of medical ethics.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


61
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

5.3.1 Pre-clinical evaluation - HSC


Step 3A: Risk reduction strategies

Testing Matrix: In vitro testing methodologies appropriate to address different risks (HPC) (please consider if the table proposed in the section 4 is
applicable)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

Test criteria Specific test

Process
qualification

Analytical
methods
related tests
In vitro
cytotoxicity
Donor cell
viability

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


62
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Donor cell
functionality

Histological
evaluation of
the ECM

Histological
evaluation of
cell content
Residual DNA
content
Biochemical
evaluation of
ECM quality

Biomechanical
properties

In vitro
functionality

Residual
processing
reagents

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


63
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

In vivo testing methodologies appropriate to address different risks (please consider if the table proposed in the section 4 is applicable)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

Test criteria Specific test

Process
qualification

Analytical
methods
related tests
In vitro
cytotoxicity
Donor cell
viability
Donor cell
functionality
Histological

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


64
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
evaluation of
the ECM

Histological
evaluation of
cell content
Residual DNA
content
Biochemical
evaluation of
ECM quality

Biomechanical
properties

In vitro
functionality

Residual
processing
reagents

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


65
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
5.3.2. Clinical evaluation and follow up plans
Step 3B: Definition of Clinical Studies

(chapters for different types of tissue)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

Test category Detailed investigational options

Physical
investigation
(discrete
outcome
measures with
quantifiable
results)

Overall Clinical
outcome
measures

Patient
Reported
outcome
measures

Procedure or
graft failure

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


66
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Post operative
complications

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


67
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

5.4. Worked examples

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


68
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

6. ART Specific Section Commented [TK17]: Can you please take care if changes have
been done on the generic explanation in the tables, that they are
copy-pasted to the specific chapters? Thank you!

There are 3 steps that need to be completed in order to determine the novelty, risks and extend
of studies needed to perform before the process is implemented in the TE.

Define which type of TCTP you are evaluating

At first it is important to define for which TCTP you are going to use the tool, as this will
generate specific risk factors. In case of ART, choose ‘Assisted Reproductive Techniques’ and
subsequently which type of reproductive TCTP is the subject of the process under evaluation.

Figure 6.1: Diagram of tool

6.1. Evaluation of Novelty (Step 1)


Before any risk analysis can be performed it has to be determined if the process change under
evaluation consists of a novelty or not. If not, then no further action is needed in addition to the
regular follow-up of established protocols. If the change in process is indeed a novelty, than the
risk assessment needs to be performed (Step 2) and the tool will determine the specifics of the
follow-up needed.

The exercise in Step 1 consists on a set of questions, which answers seek to determine if the
users are facing a novelty. Novelty is present whenever the user answers “no” to at least one of
the seven questions.

When facing all positive answers (yes), users are not dealing with something new. For this
standard/established TCP, only the regular, internal validations and follow-up procedures
should be put in place/maintained, and the level risk analysis is not required. For ease of

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


69
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

interpretation, one example is used to explain table 6.1. In this way the specificity of the
different questions is easier interpreted. The example used is: vitrification of sperm, where the
standard protocol in your TE was slow freezing.

Table 6.1: Exercise for assessing novelty

Yes No NA
A. Has this type of TCP previously been prepared and issued for clinical
use by your establishment?
Explanation: Consider if your TE has previous experience working with the TCP or not.

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question A would be YES:
you have previously prepared sperm and issued it for clinical use
B. Will the starting material used to prepare this TCP be obtained from
the same donor population previously used by your establishment for
this type of TCP?
Explanation: Consider if the starting material is from the same donor population or not.

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question B would be YES:
the starting material is from the same donor population
C. Will the starting material for this TCP be procured using a procedure
used previously by your establishment for this type of TCP?
Explanation: Consider the starting material and how it is procured or collected and if this changes in
the novel protocol or therapy.

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question C would be
YES: the starting material is procured using the same procedure. There is no change in TESE
protocol, only the cryopreservation method is different.
D. Will this TCP be prepared by a procedure (processing,
decontamination and preservation) used previously in your
establishment for this type of TCP?
Explanation: Consider the complete processing procedure of the product. If changes occur in the new
protocol or therapy, answer the question accordingly.

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question D would be
NO: there are indeed changes in processing and preservation of the sperm when vitrification will
be introduced in comparison to the standard slow freezing protocol used at the moment.
E. Will this TCP be packaged and stored using a protocol and materials
used previously in your establishment for this type of TCP?
Explanation: Consider if changes occur in the packaging and storage and if you have experience with
these items in your TE for the specific cell or tissue product where the novelty is introduced.

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question E would be
NO if there are changes in the type of packaging in the case that straws will be used for the
vitrified sperm instead of vials.
F. Will this type of TCP provided by your establishment be applied
clinically using an implantation method used previously?
Explanation: Consider if product or therapy has been clinically applied previously and answer
accordingly.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


70
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Example:
You want to implement vitrification of sperm in your TE, thus the answer to question F would be
YES: there is no difference in clinical application for the sperm
G. Has your establishment provided this type of TCP for transplantation
into the intended anatomical site before?
(this question will only appear if gonadic tissue is chosen)

Explanation: Consider the clinical application experience of the product in your TE and answer the
question concerning the intended anatomical site.

Example:
This question will only appear if gonadic tissue is chosen, hence the example for the questions above is
not relevant. For examples: This question will be answered ‘NO’ if heterotopic transplantation of
ovarian tissue strips is a new protocol, where previously only orthotopic transplantations were
performed in your clinic.

When having answered ‘NO’ to one of these question, the level of risk must be determined in
STEP 2.

6.2. Level risk analysis (Step 2)


The 2nd exercise aims to determine the risk associated with the novelties attenuated in the
process being evaluated.

Every modification in the processes associated with the donation, procurement, testing,
processing, storage and distribution of cells and tissues may have potential consequences for
the quality of these products and safety of recipients and the corresponding offspring in ART.
Moreover, different levels of novelties represent different risks and distinct impact in the quality
and safety of the tissue and cell products. The evaluation of such risks could be performed using
the methodology proposed in the current rationale.

Step 2A: Identification of risk factors

At first select the risk factors associated with the changes in the process. There are 8 factors
that could be applicable to changes in processes concerning gametes and embryos or ART
treatments and 10 risk factors that could apply to ART when gonadic tissues are concerned.
(table 6.2).

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


71
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 6.2. Identification of the risk factors Commented [TK18]: Please see my remark above concerning
the generic explanation in these tables
Consider whether the novelty in your process or
procedures changes donor characteristics and if
these changes could impart a risk to the recipient.

For example your TE is going to collect sperm from


Donation Donor Characteristics peripubertal boys (12y-14y) in comparison to the
previous protocol where only pubertal boys (>14y)
were in the donor population or anonymous sperm
donors that are being recruited from Southern
Europe where before only Northern European men
were included.
Consider where and how the TCTP is recovered
now and how this would happen when the novelty
is taken into account. Is there a change in recovery
Recovery process and
Procurement time, complexity quality of the environment?
environment

For example when a new semen container is used,


this could have an effect on the recovery process.
Consider where and how the TCTP is prepared at
the moment and how the novelty in the process
can effect this step. How long does the novel
preparation process take and how complex it is –
this may impact on the risk of contamination, or
that it may not be prepared to consistent
specifications and quality. Also consider the quality
Preparation process and of the preparation environment, which may also
environment’ affect the risk of contamination.

For example when changing from laser assisted


Processing/ storing hatching on day 3 to day 5 for trophectoderm
/transport biopsy. This change can potentially have an effect
on several risks during the preparation process. Or
changes in performing ICSI not in a laminar flow
hood, compared to doing ICSI enclosed in a hood.
Consider any reagents used during recovery,
preparation, decontamination and storage of the
TCTP. Could they damage the TCTP in any way, or
could residual traces of reagent remain in the TCTP
Reagents
that could cause toxic or immunogenic effects in
recipients?

For example when changing a cryopreservation

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


72
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

medium: this could induce a potential risk or when


using a new anaesthetic during oocyte pickup.
Consider any potential risks arising from how the
starting material and TCTP are stored, not only
after processing and before clinical application, but
also in intermediate steps: e.g. between
procurement and processing, during processing,
Storage conditions and between processing steps.

For example, when stimulation medication is now


stored at room temperature instead of refrigerated
or when sperm is being stored in liquid nitrogen and
in the novel process in vapour phase.
Consider any potential risks arising from how the
starting material and TCTP are transported, for
example between the sites procurement and
processing, and between the sites of storage and
Transport conditions clinical use.

For example, When a new type of dryshipper is


going to be used to transport frozen sperm to
clinical sites
Consider the risk that the testing methodology and
/or presence of residual processing reagents such
as antibiotics in the finished TCTP may impact the
accuracy of any microbiology/mycology testing of
the TCTP. This risk factor is not about blood tests
Reliability of microbiology on the donor.
testing (in case of gonadic
tissue)
For example, a change is processing medium of
ovarian tissue which would mask the current
microbiology testing because of the presence of
antibiotics in the new protocol in comparison to the
previous one.
Consider the risk that the changes in procedures of
processes can have on the viability or functionality
of the TCTP

Loss of viability and or


For example, change in a cryopreservation
functionality
Product processing step could result in loss of viability after
thawing or when trophectoderm biopsy is
implemented, harm to the blastocyst needs to be
taken into consideration.
Presence of donor cellular This risk must be considered from the perspective
material (in case of gonadic that for some TCTPs, the presence of intact vital

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


73
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

tissue) cells in desirable, although it may also increase the


risk of, for example immunogenicity, disease
transmission or could even lead to tumour
formation.

For example, when ovarian tissue autologous


transplantation is performed in patients with a
history of blood cancer at the moment of tissue
procurement. The risk of transmission of malignant
cells should be considered.
Vascular tissues may be more at risk to infiltration
Graft vascularity (in case of by pathogens or malignant cells than avascular
gonadic tissue) tissues.

Consider the method of clinical use for this TCTP.


Are there any novelties in this step? How long will it
take, and could this introduce risks? What is the
scope for errors to be made, and what could the Commented [TK19]: For instance also in labelling (?) Because
consequences of these errors be? this is not a risk linked to the material itself (VP)

clinical application Complexity of the application Low feasibility of application standardization might
procedure method have influence in the risks of implant failure and
disease transmission at least.

For example, when a new transfer catheter is going


to be used, is there a risk for re-transfer and how
would this affect the outcome?

Step 2B: Identification of risks

Then, when a risk factor is applicable, potential risks must be considered and the probability
scored. The potential risk must be considered in comparison with the TCTP prior to the
implementation of novelty. When selecting the potential risk, it is important to think of the
potential harm that the novelty may cause to the recipients, the resulting child and/or the
impact on the availability and accessibility of treatment. It is important to note that the risks
are not about the viability of the embryo. For example, if the viability of a blastocyst could be
harmed because of a novel biopsy procedure, than the risk factor loss of viability and/or
functionality should be chose. However the quantification of the potential risks should be
assessed bearing the recipient in mind. So, would there be unexpected immunogenicity in the
recipient when this damaged embryo would be transferred? Would there be implant failure or
pregnancy loss? Would there be a risk of disease transmission in this patient? Some examples
are given to explain the risks:
Potential risks associated with the clinical use of ART tissue and cell products are:

 Unexpected immunogenicity:

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


74
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

 This is only applicable for gonadic tissue and this option will also only appear
if gonadic tissues are selected at the start of the risk assessment. Implant
failure and/or pregnancy loss in case of gametes and embryos Commented [TK20]: I would also specifically state: loss of
batch of gametes or embryos (requiring extra treatment rounds for
 Disease transmission (including infection): the patient)
 Consider if the novelty in the TCTP has a potential risk of introducing disease Because this risk is easily forgotten (VP)

transmission or infection in the recipient. Toxicity / Carcinogenicity :


 Consider if the novelty in the TCTP can introduce toxicity reactions in
the recipient or even if there is a risk for carcinogenicity
 Other:
 Consider other risks associated with the changes in TCTP and score
them accordingly.
In order to have a complete overview of the combination of risk factors, a complete table with
examples is given. It is important to note that not all risk factors apply to changes in protocols
and procedures, likewise, not all risks apply to a risk factor. For the ease of interpretation, the
explanation of the risks is based on the example:

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


75
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Table 6.3. Combined table of the Identification of the risk factors and the associated risks

Examples and Explanation


Risk factors Risks Examples and Explanation

Unwanted
Not applicable it this example, only for gonadic tissue.
immunogenicity
Implant failure/ Consider and quantify the risk that sperm collected from peripubertal
For example your TE is going to collect sperm from pregnancy loss boys might lead to pregnancy loss when used in assisted reproduction
peripubertal boys (12y-14y) in comparison to the Although highly unlikely consider and quantify the risk that sperm
previous protocol where only pubertal boys (>14y) collected from peripubertal boys might lead to disease transmission in
Donation

were in the donor population Disease


Donor the recipient. If this population boys would be from a specific area where
transmission
Characteristics Consider whether the novelty in your process or the incidence of certain viral diseases would be known, then this might
procedures changes donor characteristics and if these impact on the risk for disease transmission.
changes could impart a risk to the recipient. It is highly unlikely that this change in donor characteristics would have a
Toxicity/Carcino risk for toxicity in the recipient. In the case of gonadic tissue that came
genicity from a donor with oncological disease, this riks must be taken into
account.
Other Consider other risks if applicable
For example when a new semen container is used, this Unwanted
Not applicable it this example, only for gonadic tissue.
could have an effect on the recovery process. immunogenicity
Implant failure/ It would be highly unlikely that the use of a new semen container during
Procurement

Consider where and how the TCP is recovered now and pregnancy loss collection would impact on implant failure.
Procurement
how this would happen when the novelty is taken into It could be possible that if this new container would be a non-sterile
process and Disease
account. Is there a change in recovery time, complexity container, that this might influence disease transmission. Although the
environment transmission
quality of the environment? risk would be rare.
Toxicity/Carcino Consider the risk that using a new semen container would have on the
genicity toxicity or carcinogenicity in the recipient.
Other Consider other risks if applicable.
Unwanted
/transpor

Not applicable it this example, only for gonadic tissue.


Processi

Preparation For example when changing from laser assisted


storing

immunogenicity
ng/

process and hatching on day 3 to day 5 for trophectoderm biopsy.


t

Implant failure/ This change in preparation will rarely effect pregnancy loss because of
environment This change can potentially have an effect on several
pregnancy loss changes in the preparation.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


76
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
risks during the preparation process. If this procedure would take place in a different environment where the
Disease
risk for environmental contamination would be higher, then a risk for
transmission
Consider where and how the TCP is prepared at the disease transmission in the recipient might be impacted.
moment and how the novelty in the process can effect Toxicity/Carcino It is highly unlikely that the change from day 3 to day 5 laser assisted
this step. How long does the novel preparation process genicity hatching would introduce toxic compounds in the recipient.
take and how complex is it – this may impact on the
risk of contamination, or that it may not be prepared to
consistent specifications and quality. Also consider the Other Consider other risks if applicable.
quality of the preparation environment, which may
also affect the risk of contamination.
Unwanted
For example when changing a cryopreservation Not applicable it this example, only for gonadic tissue.
immunogenicity
medium Implant failure/
The change in reagents will unlikely impact on the risk of pregnancy loss.
pregnancy loss
Disease If this new reagent contains for example albumin from a source that is
Reagents Consider any reagents used during recovery, transmission doubtful, then there is a risk for disease transmission to the recipient.
preparation, decontamination and storage of the TCP. Toxicity/Carcino If this medium contains different types of antibiotics, than this might
Could they damage the TCP in any way, or could genicity have an impact on toxicity reactions in the recipient.
residual traces of reagent remain in the TCP that could
cause toxic or immunogenic effects in recipients? Other Consider other risks if applicable

When sperm is being stored in nitrogen vapour phase Unwanted


Not applicable in this example, only for gonadic tissue
rather than liquid nitrogen. immunogenicity
Implant failure/ The change is storage conditions might have a direct impact on implant
pregnancy loss failure when this preserved sperm would be used for insemination.
Consider any potential risks arising from how the
Storage Disease The change in storage might theoretically have an impact on disease
starting material and TCP are stored, not only after
Conditions processing and before clinical application, but also in transmission transmission, even though the risk is very rare.
intermediate steps: e.g. between procurement and Toxicity/Carcino The impact of the novel storage conditions will, in this example have very
processing, during processing, and between processing genicity little, even no impact on the introduction of toxic compounds.
steps.
Other Consider other risks if applicable

Transport Unwanted
For example, when an ART centre is starting IVF in Not applicable it this example, only for gonadic tissue.
Conditions immunogenicity

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


77
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
transport or satellite conditions where oocytes are Implant failure/ New transport conditions might have an impact on pregnancy loss if not
collected in another clinic. pregnancy loss adequately controlled.
Disease Disease transmission is rarely impacted if only transport conditions are
Consider any potential risks arising from how the transmission changed.
starting material and TCP are transported, for example Toxicity could be impacted if not only transport conditions are changed,
Toxicity/Carcino
between the sites procurement and processing, and but maybe medium differences are also present between the satellite
genicity
between the sites of storage and clinical use. center and the current TE.
Other Consider other risks if applicable
For example, a change in processing medium of Unwanted This change could have an impact on unwanted immunogenicity when
ovarian tissue which would mask the current immunogenicity this tissue is transplanted in the recipient.
microbiology testing because of the presence of Implant failure/ This change could lead to implant failure due to residual microbiological
Reliability of antibiotics in the new protocol in comparison to the pregnancy loss load that has an impact on the graft viability.
Microbiology previous one. If this change in solely on processing medium, but it is still autologous
Disease
Testing use of the tissue, then the risk of disease transmission will probably not
transmission
(in case of chance in comparison to the former procedure.
gonadic tissue) Toxicity/Carcino
Consider the risk that the testing methodology and /or There might be a risk for introducing toxic compounds.
genicity
presence of residual processing reagents such as
antibiotics in the finished TCP may impact the accuracy
of any microbiology/mycology testing of the TCP. This Other Consider other risks if applicable
risk factor is not about blood tests on the donor.
Unwanted
For example, change in a cryopreservation processing Not applicable it this example, only for gonadic tissue
immunogenicity
step could result in loss of viability after thawing or Implant failure/ This novelty can have a direct impact on implant failure and pregnancy
Loss of viability when trophectoderm biopsy is implemented, harm to pregnancy loss loss.
and or the blastocyst needs to be taken into consideration. Disease The impact on disease transmission due to harming of the embryo
Product

functionality transmission because of the new biopsy technique is highly unlikely.


Consider the risk that the changes in procedures of Toxicity/Carcino The loss of viability will probably only have a rare impact on the
processes can have on the viability or functionality of genicity introduction of toxicity or carcinogenicity in the recipient.
the TCP
Other Consider other risks if applicable
Presence of Unwanted Consider the risk that presence of cellular material could have on
donor cellular immunogenicity unwanted immunogenicity in the recipient.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


78
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
material For example, when ovarian tissue autologous Implant failure/ There could be a risk for implant failure when malignant cells are present
(in case of transplantation is performed in patients with a history pregnancy loss in the graft.
gonadic tissue) of blood cancer at the moment of tissue procurement. Disease If malignant cells are transplanted together with the graft, the there is a
The risk of transmission of malignant cells should be transmission risk for the transmission of oncological disease.
considered. Toxicity/Carcino
Presence of cells might impact on the risk of carcinogenicity
genicity
This risk must be considered from the perspective that
for some TCPs, the presence of intact vital cells in
desirable, although it may also increase the risk of, for Other Consider other risks if applicable
example immunogenicity, disease transmission or
could even lead to tumour formation.
Unwanted Score the risk that the graft vascularity might have on unwanted
immunogenicity immunogenicity in the novel procedure
Score the risk that the graft vascularity might have on implant failure in
Graft Implant failure
the novel procedure
vascularity Vascular tissues may be more at risk to infiltration by
Disease Score the risk that the graft vascularity might have on disease
(in case of pathogens or malignant cells than avascular tissues.
transmission transmission in the novel procedure
gonadic tissue)
Toxicity/Carcino Score the risk that the graft vascularity might have on toxicity in the
genicity novel procedure
Other Consider other risks if applicable
Unwanted
Not applicable it this example, only for gonadic tissue
For example when a new transfer catheter is going to immunogenicity
Clinical application procedure

be used, is there a risk for re-transfer and how would Implant failure/ There might be an impact on the risk of implant failure when a new
this affect the outcome? pregnancy loss transfer catheter is introduced.
Complexity of Disease It is highly unlikely that the risk for disease transmission would be
the Consider the method of clinical use for this TCP. Are transmission impacted when only a new transfer catheter is implemented.
preparation/ap there any novelties in this step? How long will it take,
plication and could this introduce risks? What is the scope for Toxicity/Carcino A new transfer catheter might, although unlikely, introduce toxicity to
method errors to be made, and what could the consequences genicity the recipient
of these errors be?
Low feasibility of application standardization might
have influence in the risks of implant failure and Other Consider other risks if applicable
disease transmission at least.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


79
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Step 2C: Quantification of risks


When the risk factors are selected and the potential risks are identified, the potential impact of
this risk analysis needs to be determined:
i) What is the probability that the Impact will occur:
Table 6.4. – Probability levels9
Level of probability Definition
1 - Rare Difficult to believe it could happen
2 - Unlikely Not expected to happen but possible
3 - Possible May occur occasionally
4 - Likely Probable but not persistent
5 - Almost certain Likely to occur on many occasions

ii) What is the severity of the outcome, should the impact occur:

Table 6.5 – Severity levels10


Level of
Definition
severity
Non-serious Mild clinical or psychological consequences. No hospitalisation, or anticipated long
term consequences/disability
Serious Hospitalisation and/or:
 Persistent/significant disability or incapacity
 Intervention to preclude permanent damage
 Evidence of a serious transmitted infection
 Significant decrease of expected treatment success (e.g. total loss of gametes
or embryos)
 Significant risk of miscarriage
 Birth of a child with a disease following ART with donor gametes or embryos
Life-  Major intervention necessary to prevent death
threatening  Evidence of a life threatening transmissible infection
 Birth of a child with life threatening genetic disease related to ART with donor
gametes of embryos
Fatal Death of the patient

iii) What is the detectability in the likelihood of the impact occurring


The detectability is the chance for the potential effect to be detected” in a certain time frame
before there is an effect in the recipient or child. It refers to the probability of detection of the
potential effect before producing any harm.
Classification of detectability:

9
Definitions from V&S SoHO Project, 2009 [10]
10
Definitions from EUSTITE, 2009

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


80
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 6.6. – Detectability levels


Level of detectability Definition
The potential defect/harm will almost certainly be detected before
clinical application in the recipient (for example: embryo of very
poor quality due to the new processing method and is therefore not
1 - Very high
eligible for transfer). ). (This means almost certainty about the
potential effect)

There is a reasonable chance that the potential defect/harm will be


2 - Moderately high
detected before clinical application in the recipient
There is a low chance that the potential defect/harm will be
detected before clinical application in the recipient (potential
3- Low
defect/harm requires additional quality controls and may not be
detected by the current available techniques)
It is unlikely that the potential defect/harm will be detected before
clinical application in the recipient (for example, it could be that
4 - Very low
there are epigenetic changes in the embryo that cannot be
determined without destroying the embryo).
The potential defect/harm will be detected only after clinical
application in the recipient (for example, an increased possibility of a
5 - Cannot be detected child having an abnormality or pathology due to the novelty). (This
means absolute uncertainty about the potential effect)

iv) To what degree can this risk be reduced by analysis of pre-existing data11
Risk reduction can be take into account when there is data evidence that supports reducing the
overall risk score. This may be (for example) published data in the peer reviewed literature,
internal validation reports, clinical outcomes, or any other relevant sources of data. The quality
of the data should also be taken into consideration, for example data in a high impact factor,
peer reviewed publication should be given more weight than unpublished data.
The risk estimated when implementing novelties may be reduced by:

 Bibliographic references that demonstrate safe and efficacy of products and/or


techniques;
 Technical improvements that may decrease the risk associated with the activities (e.g.:
when the novel process represents a change of processing method from “open system”
to a “close system”, the risk factor “time of exposure” may represent a reduction of
risks when processing with the novel method.)
Based on your evaluation of the totality of the evidence, different levels of risk reduction
may be assigned according the scale:

11
Pre-existing data is defined as data generated either by your organisation or by others that indicates
the probability or severity score may be lower than the score you have selected, or that the likelihood of
detection may be higher than the score you have selected.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


81
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Table 6.7. – Percentage risk reduction definitions


Percentage
risk Definition
reduction
There is no relevant data available to support reducing the calculated risk
0
score
There is a moderate relevant data available to support reducing the calculated
25
risk score, based predominantly on unpublished, internal data
There is good quality relevant data available support reducing the calculated
risk score, including published and unpublished data from internal and
50
external sources, and some data which has been through an independent peer
review process
There is high quality relevant data to support reducing the calculated risk
75
score, including data that has been peer reviewed and published.
There is a substantial amount of high quality relevant data, including multiple
95 peer reviewed publications, that demonstrates that the probability of the risk
occurring, having a significant impact, and/or being undetected is negligible.

In order to provide you with a comprehensive overview of the complete risk assessment of 1
particular example, see attachment x. In this example, rather than in the explanation above, a
complete exercise for 1 novelty is shown. This can aid in the comprehension of the above
descriptions.

6.3. Definition of extent of studies needed based on the risks quantified (Step 3)

Using the tool you will be able to perform a risk analysis, determine the risk profile and the level
of risk associated with the novel process or procedure. As a result the tool will provide the total
risk value (number) and the final risk value which is proportional to the number of risk
evaluated (in the form of a level of risk). It is important to state that ART centres should be
prepared to invalidate treatment when proven problematic (in terms of safety and
effectiveness) even when a novelty of negligible risk was implemented. ART centres should
collect data and register of follow-up in a systematic way and make them available to the
scientific community regardless of the success of the treatment: not withholding results that
point to a negative outcome or that turn out to be inconclusive (Provoost V. et al. 2014).
Therefore it is important in all processes, regardless of the level of risk, to monitor and register
SARE / SAE.

The table below gives guidance on the follow-up studies needed in term of the level of risk
determined (adjusted according to Provoost V. et al. 2014).

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


82
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Level of Risk Extend of Studies needed


A change in process could have a negligible level of risk because it is part of a
therapy or procedure that is considered as established or standard.

In this case multi-centred studies (ideally RCT) are published in peer-reviewed


journal and the procedures are performed according to a validated and standard
protocol. Minimal process validation is needed. The technical performance of staff
should be monitored and comparable with other TE or published studies, therefore
standard Key Performance indicators (KPI) should be monitored on the technical
Negligible quality of the staff performing the procedures. Dropping KPIs indicating protocol
drift must lead to investigation of both the procedural steps and / or the possibility
to re-train staff.

A routine/safety follow up program is enough as the good practices state. Follow-up


procedures should be focused on assessing efficacy, comparing the clinical follow-
up with the results obtained before the implementation of the change in the
process. Long-term (ideally trans-generational) health effects, including aspects
such as fertility, oncology and mental health should be monitored.

Implementing a standard procedure or treatment in an ART centre that has never


performed this procedure exerts an intensive validation. Training of staff is
necessary in order to reach the outcomes published in scientific literature. Having
a mentor/mentee relationship with an ART centre having experience is highly
recommended. Specifications on performance should be determined and when
these limits are met by training on spare tissues and cells, staff can be authorized
for performing the procedure.

A learning curve might be expected and should be part of the validation report.
When implementing the procedure, additional quality controls must be performed
to monitor Critical Process Parameters (CPPs) and Critical Qualtiy Attributes (CQAs).
For example, when a TE is switching from IVF to ICSI (which they never performed
before), fertilisation rated, and damage rates etc. of embryos should be carefully
Low monitored in relation to the staff performing the procedure.

A safety follow up program is necessary. Follow-up procedures should be focused


on assessing efficacy, comparing the clinical follow-up with the results obtained
before the implementation of the change in the process and in relation to the
results published in scientific literature. As the procedure or treatment
encompasses an established or standard technique. The expected learning curve
should be kept as short as possible and put in relation to the follow up program.

Likewise, established techniques are prone to long-term (ideally trans-generational)


follow-up of the health effects. TE or ORHA implementing an established technique
shall perform long-term follow-up and could base their follow-up items on the
mentor facility. This way of working could lead to periodic evaluation of
performance in the mentor/mentee relationship.

Novel procedures or treatments that exert a moderate risk and are considered
Moderate innovative. The treatment has shown proof of principle and there is reassuring data
in literature in terms of both safety and effectiveness at least in animal studies and

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


83
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

pre-clinical data shows normal embryology development. The studies that have
published this data should have a sound methodology and published in peer-
reviewed journals.

In order to implement an innovative treatment, an enhanced validation is necessary


including and a range of additional quality controls performed to monitor Critical
Process Parameters (CPPs), Critical Quality Attributes (CQAs), and the impact of the
implemented changes on gametes, embryo’s and gonadic tissue should be carefully
monitored. Since reassuring data of this innovative treatment is already available, a
more specific monitoring of the published critical parameters can be performed
instead of a registration of all critical parameters.

Clinical evaluation and follow up programs should be implemented to assess


reassuring mid-term safety (3 months up to 5 years post-delivery including data on
psychological wellbeing) and these studies should refer to patients undergoing the
procedure as well as the children born from it.

A new procedure can be offered to patients in an experimental design aiming at


showing proof of principle, short-term safety and/or effectiveness.

An extensive validation including and a range of additional quality controls


performed to monitor Critical Process Parameters (CPPs), Critical Quality Attributes
(CQAs), and the impact of the implemented changes is required. This extensive
validation should include:

Non clinical studies: preferably there should be studies showing the experimental
procedure is safe in animals.

Pre-clinical Studies: when experimental treatments encompass a laboratory IVF


High phase, then at least the structural integrity of the gametes, embryos or gonadic
tissue should be looked at in detail, monitored and registered. Clinical embryology
data should indicate a normal cleavage embryo morphology and blastocyst
formation.

Follow up program: experimental treatments should only be offered to a selected


and limited patient cohort and these patients should be clearly informed on the
experimental status and should receive information about (the lack of knowledge
about) possible risks, alternative treatments etc. ORHAs should only offer
experimental treatments or treatments based on experimental procedures after
approval by a commission of medical ethics.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


84
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

6.3.1 Pre-clinical evaluation - ART


Step 3A: Risk reduction strategies

Testing Matrix: In vitro testing methodologies appropriate to address different risks (ART)

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

procurement or
acquired during
Genetic disease
Carcinogenicity

in the offspring
Pregnancy loss
Implantation

Blood borne
Anaphylaxis

cytotoxicity

processing
Infections
infections
Test criteria Specific test
Localised

Localised
response

response
Systemic
Immune

immune

failure
Sterile handling

Morphology
testing

Vitality
Fertilization
rate
Process
Morphokinetics
qualification
Blastocyst
formation rate
Materials and
reagents
quality specs
definition
Packaging
sealing test
Validation of
Analytical the virology &
methods microbiology
related tests analytical
method

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


85
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Validation of
the vitality
testing
Validation of
morphology
related scoring
systems
In vitro N.a.?
cytotoxicity N.a.?
Microscopy
observation
Donor cell Metabolic
viability activity assays
Live/dead
staining
Microscopy
observation
Donor cell Quantification
functionality of secreted
factors
?
H&E stain
Immunohisto-
Histological
chemical
evaluation of
testing
gonadal tissue
Apoptosis
testing
?
DNA integrity
testing

DNA content PGS

PGD
Semen fructose
levels
Biochemical Semen Zn
evaluation levels
Hormone levels

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


86
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Sperm 24 hours
survival test
In vitro
HZA
functionality
Steroidogenic
activity
HPLC
Mass
Residual spectrometry
processing Biochemical
reagents tests
Immunohisto-
chemistry

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


87
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
In vivo testing methodologies appropriate to address different risks

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

procurement or
acquired during
Genetic disease
Carcinogenicity

in the offspring
Pregnancy loss
Implantation

Blood borne
Anaphylaxis

cytotoxicity

processing
Infections
infections
Test criteria Specific test

Localised

Localised
response

response
Systemic
Immune

immune

failure
Implantation
rate
Pregnancy
rate
Miscarriage
rate
Biocompatibility Delivery rate
??? Resumption
of regular
menstruation
Resumption
of ovulation
?
?
Blood count
Leucocyte
Immunological immunophe-
response notyping
Local
immunology
testing
US doppler
Thrombotic
Coagulation
response
testing
US scans
Hormone
Functionality
levels
?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


88
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
Tumor
In vivo markers
carcinogenicity MRI
Laparoscopy
± biopsy
General
wellbeing
OHSS
Health Multiple ? ? ? ????? ? ? ? ? ? ?
pregnancy
Infection
Ovulation
Reproductive detection
function Pregnancy
restored detection
?
Prematurity
(g.w. at birth)
Birth weight
Congenital
Offspring defects
Mental
wellbeing
?
?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


89
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
6.3.2. Clinical evaluation and follow up plans
Step 3B: Definition of Clinical Studies TBC

Immunogenicity Graft failure Toxicity/Carcinogenicity Disease transmission

Test category Detailed investigational options

Physical
investigation
(discrete
outcome
measures with
quantifiable
results)

Overall Clinical
outcome
measures

Patient
Reported
outcome
measures

Procedure or
graft failure

Post operative

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


90
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))
complications

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


91
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

6.4. Worked examples

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


92
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

7. Tissue & Cell Database


The Tissue and Cells (T&C) database aims to be compendium of tissues/cells products,
preparation processes, applications and therapies.
7.1. Purpose of the T&C Database
The purpose of the European T&C database is to promote the safe and effective use of Tissue
and Cellular Therapies/Products (TCTPs), by the provision of data related to the products and
therapies available, and references relating to their efficacy.
The structure and contents of the database were defined in order to ensure its consistency,
harmonize the characterization of TCTPs, and support the collection of efficacy and quality data
associated with the clinical use of Substances of Human Origin (SoHO) at European level.
The T&C Database was designed to be appropriate for the needs of:
- Tissue Establishments (TEs) and those engaged in the quality control and design of pre-
clinical studies and clinical evaluation of TCTP;
- Users / Organisations Responsible for Human Application (ORHAs);
- Competent Authorities (CAs);
- Patients;
The distribution of TCTP between European Member States is a common practice, and the
exchange of scientific and clinical information promotes the assessment of safety and efficacy of
novel and traditional SoHO’s therapies.
The aim of this tool is to provide structured and systematic information regarding TCTPs
implemented by the TEs, and include an overview regarding new TCTPs, information on clinical
application and references to available efficacy and safety data .
This information contained in the T&C Database intends to:

- Collate references and evidence relating to safety and efficacy data;


- Encourage stakeholders/CAs to accept the validity of data generated for products in
other countries (harmonization of practices)
- Promote collaboration amongst TEs, encouraging multicenter collaborations for the
development of novel TCTPs;
- Promote the accessibility of TCTP for patients.
The data included in the T&C Database was voluntarily shared by Tissue Establishments, with
the intention of contributing to the knowledge base associated with novel and well established
TCTPs within Europe.
This data should be periodically reviewed and updated by experts nominated by the European
scientific associations that collaborated with the EuroGTP II Project: EBMT, EATB, EEBA and
ESHRE. This review aims to ensure that the data is trustworthy and up to date, and to avoid
redundant entries.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


93
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

7.2. General Principles


The T&C Database is a registry of TCTP consisting of information provided by European TEs.
TEs are encouraged to register information associated with clinical evaluation studies
performed to determine the safety and efficacy of the TCTP distributed.
The information provided will assist stakeholders in the development of pre-clinical studies and
clinical evaluation of novel TCTPs. Furthermore, this information promotes dialogue between
the European CAs and TEs seeking collaborations and sharing of expertise and information.
In order to assure consistency and scientific reliability, the EuroGTP II project has defined the
principles and procedures required for the correct inclusion and interpretation of data
submitted to T&C Database.
The technical guidelines (instructions and definitions) to correctly complete, submit and review
data, are part of the current document and intend to assist users and contributors.
The principles applied should be periodically revised by experts in according the proposed in the
“GTP’s Management Model”. (???)
TEs contributors should provide sufficient information to ensure that the data included is
robust, comprehensive and evidence based. Data relating to authorisation status and validation
supporting the TCTP (preparation process?) should also be provided.
Each record includes a summary description and information about the current status of the
TCTP with regard to clinical uses, risks assessed and authorizations status
Some TCTP entries may include the number of recipients already treated on an annual basis, the
number of patients defined for the clinical evaluation studies, and cross references to the Notify
Library.

7.3. Introduction of data:


The methodology for data entry, data review and data management still needs to be defined.
Scientific associations will definitely play a critical role in the future management of the
Database.
Our goals is to have the products issued by all TEs partners in the T&C Database.
A very simple prototype form for the introduction of data is available online – here.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


94
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

7.4. Description of Contents


M – Mandatory field | OP – Optional field | FA – Filled automatically in the future

TE Description
M EU TE Code EU TE Code (SEC - - 2 letters 6 Numbers)
M TE Name Full Name of TE
Name of Country + ISO code (2 letters code
M Country
of ME)
OP Website/other info www. Not yet included Commented [a21]: Answer to NHSBT’s comment

The information related with TE will be directly imported from SEC


platform
Product Description Accredited source of info by NCAs)

M Version Date dd/mm/yyyy (will be generated automac.) Authorised activities should be part of this info

Some infos were deleted in the online form to simplify the


M Product ID EUTC Code (Select Option) introduction of data in the prototype (limited number of fields
available)

M SoHO Class Tissue/Cells/ART (Select Option) Commented [LR22]: Potential other fields within this category:
1.Status of the organisation, e.e. public/private sector
Amniotic Membrane/Cardiovascular/Ocular/Other 2. Which aspects of the TCTP supply chain does the organisation
M Product Type Membranes/Mature cells/MSK/Progenitor Cells/ Skin/ Embryo perform itself
3. How long has the TE been established
/Oocyte /Ovarian Tissue/ Sperm /Testicular Tissue (Select Option) 4. What accreditations/licences does the TE hold?
Open text optional 5.
OP Product Sub classification
Example: Tendon/ligament/ cartilage/ etc… Commented [LR23]: Potential other fields within this category:
6.Status of the organisation, e.e. public/private sector
M Product Name Open text - (Product name given by the TE) 7. Which aspects of the TCTP supply chain does the organisation
perform itself
Open text - (Main characteristics/specifications of the product, 8. How long has the TE been established
M Product Characteristics
defined by the TE) 9. What accreditations/licences does the TE hold?
10.
Commented [LR24]: This could be subdivided into bone,
tendón/ligament and cartilage
Process Description
[RP]This will be considered under “product sub classification”
Allogeneic (postmortal donors)/
Donor/Recipient Allogeneic (living donors)/ related/unrelated
OP
Relationship Autologous
(Select Option)
Open text (Optional)
OP Specific Donor Criteria (Donor selection criteria applied, over and above EUCTD
requirements)
Default
Collection/Recovery Ejaculated
OP
Method Extracted
(optional only for ART)
Describes additives introduced during the processing of the product.
OP Additive Solution
Text (optional)
No pathogen reduction / Not specified / Antibiotics / Combined
OP Pathogen Reduction process / ETO / No pathogen reduction / Peracetic acid / Radiation
sterilization/ Other (optional)
OP Storage Solution

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


95
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Not specified/default / Cryopreserved / Dehydrated / Freeze dried /


OP Preservation Frozen / Glycerol (high conc) / Refrigerated / Solvent dehydrated
(optional)
Other Info: (Storage
Temperature; Storage
OP requirements after issue Open text (Optional)
and/or Shelf life from
donation/after issue)
Update (innovation and
OP
changes)

Indication

OP Classification of Diseases Code (1 letter + 2 digits) - Optional

Detailed ICD10 Code (Codes: http://apps.who.int/classifications/icd10/browse/2016/en -


OP
(Disease Classification) Optional)
Result given by EuroGTP II IAT - Evaluation made by TEs
M Level of Risk - IAT Level Select from: Negligible/low; moderate; high; Not performed
(implemented before EuroGTP II)
When was the risk assessment performed - DD/MM/YYYY
M Risk Assessment Date
Not yet included in the prototype
Links or References
OP Bibliographic References Attach papers to the database
(Submission of open Access references Commented [a25]: I think this functionality can be available in
the future DB. I am not able to implement this in the prototype
OP Notify references Relevant Codes of Notify Library or Links

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


96
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

7.5. Codes used:


 EU TE ID codes – here
 Product ID Code –SEC Platform
 Classification of Diseases - Here
 Notify Library – Here

7.6. Structure of Data


Database Scheme / Entity Relationship Diagram

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


97
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

7.7. Queries and Practical Use of the database:

As mentioned above, the database may be used to search for products and therapies made
available by different TEs in Europe.

In principle, one TE can register several different TCTPs, and the same TCTPs can be prepared
and distributed by several different TE in Europe.

Different users may find it useful to perform different queries depending on their interests and
goals. Examples:

 TEs searching for collaborations may want to know who in Europe is preparing a
particular TCTP in order to establish a collaboration or gather scientific references;
 When CAs intent to search for references of TCTPS previously authorised in other
Member States, but implemented for the first time by national TEs;
 Surgeons may search for new TCTP options to treat specific pathologies;

These examples were used to validate the functionality of the T&C Database.

Definition of queries:
By TEs - Output:

 Information about the TE and


 List of TCTP registered by the TE: the user should select the
product (amongst the ones listed/TE) to access the information
related with Product ID, Experience and Authorisation.

By Product – Output:

 Product details, and


 List of TEs that currently issue/distribute this TCTP: If user
selects one of TEs that distribute the product the details
associated with Experience and authorization become
available.

By Disease or Clinical procedures - Output:

 List of TCTP registered for the selected pathology


 User may select one TCTP (and follow product query)

(...)

7.8. Management of Contents and Certification Procedures


(...)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


98
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Committee of Scientific Associations Board?

New GAPP joint Action?

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


99
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Bibliografía

[1] “DIRECTIVE 2004/23/EC OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL on setting
standards of quality and safety for the donation, procurement, testing, processing,
preservation, storage and distribution of human tissues and cells”.

[2] “COMMISSION DIRECTIVE 2006/17/EC, implementing Directive 2004/23/EC of the


European Parliament and of the Council as regards”.

[3] “COMMISSION DIRECTIVE 2012/39/EU, amending Directive 2006/17/EC as regards certain


technical requirements for the testing of human tissues and cells”.

[4] “COMMISSION DIRECTIVE (EU) 2015/565, amending Directive 2006/86/EC as regards


certain technical requirements for the coding of human tissues and cells”.

[5] “COMMISSION DIRECTIVE (EU) 2015/566, implementing Directive 2004/23/EC as regards


the procedures for verifying the equivalent standards of quality and safety of imported
tissues and cells”.

[6] Commission Directive 2006/86/EC of 24 October 2006, implementing Directive 2004/23/EC


of the European Parliament and of the Council as regards traceability requirements,
notification of serious adverse reactions and events and certain technical requiremen.

[7] E. Project, Euro GTP Guidance (Project 2007207), 2011.

[8] ARTHIQS -Assisted Reproductive Technologies and Haematopoietic stem cells


Improvements for Quality and Safety throughout Europe, Deliverable 9 “Guide of
Recommendations for Cord Blood Banking.

[9] ARTHIQS -Assisted Reproductive Technologies and Haematopoietic stem cells


Improvements for Quality and Safety throughout Europe, Deliverable 8 “Guidance for
Establishing a Hematopoietic Progenitor Cells Donor Follow-Up Registry.

[10] (. N. 2. Vigilance and Surveillance of Substances of Human Origin (SOHO V&S), SOHO V&S
GUIDANCE FOR COMPETENT AUTHORITIES:COMMUNICATION AND INVESTIGATION OF
SERIOUS ADVERSE EVENTS AND REACTIONS ASSOCIATED WITH HUMAN TISSUES AND
CELLS, 2013.

[11] European Union Standards and Training for the Inspection of Tissues Establishments
(EUSTITE) - Project 2005204, 2009.

[12] E. D. f. t. Q. o. M. (. Council of Europe (CoE), Guide to the Quality and Safety of Tissues and
Cells for Human Application, 3rd edition, 2017.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


100
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

[13] FACT-JACIE, International Standards for Hematopoietic Cellular Therapy Product


Collection, Processing, and Administration, 2015.

[14] ESHRE, Guideline of the European Society of Human Reproduction and Embryology -
Revised guidelines for good practice in IVF laboratories, 2015.

[15] E. T. B. V. N.-d. F. G. R. G. S. J. o. b. o. t. W. G. M. o. t. W. M. D. A. Van Walraven SM,


BBMT, 2018.

[16] R. JD., “Basic ethical principles in European bioethics and biolaw. Authonomy, dignity,
integrity and vulnerability – Towards a foundation of bioethics and biolaw.,” Medicine,
Health Care and Philosophy, 2002;, vol. 5, pp. 235-244, 2002.

[17] S. Z., “Right-to-Try investigational therapies for incurable disorders.,” Continuum


(Minneap Minn), vol. 23(5), pp. 1451-1457, 2017.

[18] C. A., “Medical Ethicist Arthur Caplan Explains Why He Opposes ‘Right-To-Try’ Laws.,”
Oncology, vol. 30(1):8, 2016.

[19] M. DE, “Ethical aspects of medical products of human origin.,” ISBT Science Series, vol. 12,
pp. 281-287, 2017.

[20] “https://www.eurordis.org/,” 28 February 2018. [Online].

[21] A. N. V. d. V. S. Bunnik EM, “The changing landscape of expanded access to investigational


drugs for patients with unmet medical needs: ethical implication.,” Journal of
Pharmaceutical Policy and Practice, p. 10:10, 2017.

[22] B. M., “Stem cell research: time for a dose or realism. BMJ,” BMJ, p. 356:j443, 2017.

[23] R.-S. E. Blassime A, “Regulation of Cell-Based Therapies in Europe: Current Challenges and
Emerging Issues.,” Stem Cells and Development, vol. 22(S1), p. 14:19, 2013.

[24] S. A. B. H. G. M. D. P. K. N. L. P. M. E. M. M. N. A. R. A. R. d. E. C. S. J. P. J. A. J. N. D. S. T. B.
R. Gratwohl A, “Economics and Outcome after hematooeitic stem a retrospective cohort
study. for the Joint Accreditation Committee (JACIE) of the International Society for
Cellular Therapy (ISCT) and the European Society for Blood and Marrow Transplantati,”
EBioMedicine , vol. 2, pp. 2101-2109, 2015.

[25] R. M. N. S. W. W. Elie Akl, GRADE Handbook, GRADE Working , 2013.

[26] B. M., “Stem cell research: time for a dose or realism.,” BMJ, p. 356:j443, 2017.

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


101
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


102
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Annex I – Partners and Experts of EuroGTP II Project


Associative Partners
Coordinators – Banc Sang i Teixits (BST) Esteve Trias
Leaders WP1, 4 and 9 www.bst.cat Jaime Tabera
Rita Piteira

BST Experts and Staff:


Anna Vilarrodona
Elba Agustí
Elisabet Tahull
Eva Maria Martinez
Ivan Miranda
Maria Luisa Perez
Marta Torrabadella
Nausica Otero
Oscar Fariñas
Ricardo Casaroli
Sergi Querol
National Health Service – Blood and
Akila Chandrasekar
Transplant (NHSBT)
Richard Lomas
www.nhsbt.nhs.uk
WP2 Leader Organización Nacional de Trasplantes Mar Carmona
(ONT) Esteban Molano
www.ont.es Myriam Ormeño
WP3 Leader Ministry of Health of the Republic of
Marijana Dragović
Croatia (MZRH ) - Institute for
Transplantations and Biomedicine
www.kbc-zagreb.hr
Branka Golubić Ćepulić
In collaboration with
Ivan Rozman
Klinički Bolnički Centar Zagreb (KBCZ)
WP5 Leader Italian National Transplant Centre (ISS- Cristina Pintus
CNT) Eliana Porta
www.iss.it | www.trapianti.net Fiorenza Bariani
Letizia Lombardini
Liliam Santilli
Mariapia Mariani
Paola Di Ciaccio
Silvia Pisanu
WP6 Leader Krajowe Centrum Bankowania Tkanek i Artur Kamiński
Komórek (KCBTiK) Izabela Uhrynowska-Tyszkiewicz
www.kcbtik.pl Ewa Olender
WP7 Leader TRIP Foundation, Netherlands office for Arlinke Bokhorst
hemo- and biovigilance (TRIP) Anne Marie van Walraven
www.tripnet.nl Ingrid van Veen
WP8 Leader Ghent University Hospital (UZGent) –
Kelly Tilleman
Department of Reproductive Medicine

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


103
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

www.uzgent.be Tolpe Annelies


Veerle Provoost
Lieve Nuytinck
Other Associative Partners Bulgarian Executive Agency for Maryana Simeonova
Transplantation (BEAT) Daniela Staneva-Petkova
www.bgtransplant.bg Dessislava Tzoneva
Tsvetelina kircheva-Nikolova
Violetta Marinkova
Valery Georgiev
Yoran Peev
Elizabeth Manova
Semmelweis University, Health Services Éva Belicza
Management Training Center, SU Judit Lám
(HSMTC) Gábor Szarvas
www.semmelweis.hu Cecilia Surján
László Bencze
German Society for Tissue Martin Börgel
Transplantation GmbH (DGFG) Mareike Derks
www.gewebenetzwerk.de Sibylla Schwarz
Saint Jean Clinic, European Homograft Ramadan Jashari
Bank (CSJ/EHB) Richard N. Noumanje
www.klstjan.be Rosario Daiz Rodriguez
Regea Cell and Tissue Center, University Tiia Tallinen
of Tampere Hanna Kankkonen
www.regea.fi/en Toni-Karri Pakarinen
Ecole Royale Militaire – Koninklijke Gilbert Verbeken
Militaire School (ERM/KMS) Jean-Paul Pirnay
www.rma.ac.be/en/ Thomas Rose
Collaborative Partners European Association of Tissue Banks
Simone Hennerbichler
(EATB)
Jill Davies

European Society of Human Cristina Magli


Reproduction and Embryology (ESHRE) Nathalie Vermeulen
Monserrat Boada
The European Society for Blood and
Eoin McGrath
Marrow Transplantation (EBMT)
European Eye Bank Association (EEBA) John Armitage
Gary Jones
EDQM, CoE - European Directorate for
the Quality of Medicines & HealthCare, Marta Fraga
Council of Europe
Instituto Portugues do Sangue e da
Dulce Roldao
Transplantaçao (IPST,IP)
Josefina Oliveira
www.ipst.pt
Fondazione Banca dei Tessuti di Treviso Adolfo Paolin

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


104
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Onlus Diletta Trojan


www.fbtv-treviso.org
Fondazione Banca degli Occhi del Veneto
Diego Ponzin
Onlus
Stefano Ferrari
www.research.fbov.org
Rome La Sapienza University Francesco Lombardo
Gruppo Italiano per il Trapianto di
Midollo Osseo,cellule staminali
emopoietiche e terapia cellulare
Sanquin Blood Supply Foundation Carlijn Voermans
ETB-BISLIFE
Invited Experts Leiden University Hospital Tanja Netelenbos
Hungarian Stem Cell Donor Registry at
the National Hungarian Blood Katalin Rajczy
Transfusion Service
University of Warsaw Grezgorz Basak
Ghent University Hospital (UZGent) Helene Schoenmans
Karolinksi Instituut Stockholm Stephan Mielke
University Hospital Center Zagreb Ines Bojanic
Institut Paoli Calmettes Cell Therapy
Boris Calmels
Facility
University of Oulu Zdravka Veleva
Nij Geertgen, Centre for Fertility Martine Nijs
MC ReproBioMEd Gueorgui Nikolov
External Auditors Centro Hospitalar do Porto
Margarida Amil
www.chporto.pt
Irish Blood Transfusion Service
Sandra Shaw
www.ibts.ie
Other Notify Project
Aurora Navarro
Contributors/Collaborators www.notifylibrary.org
(Including open
Consultation)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


105
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Annex II – Template Form: Characterization of TCTP

Name/description of the TCTP

Tissue Establishment ID

Confidentiality Statement TEs may decide to treat the document as confidential, if the
novelty status of the TCTP demands it.
In this situations, the Characterization form should inform that
the included information should be disclosed only within a
specific department/organisation.

Brief summary Highlighting the significant proprieties of the TCTP and/or


clinical applications under study main differences with related
SoHO products.

TCTP Characterization Main characteristics of the TCTP/Critical attributes of TCTP.


Example: (number of cells, structural characteristics), origin of
TCTP (autologous/allogeneic), excipients or other reagents or
residues could be transplanted with the TCTP (such as carriers
or preservants), and description of novelty (if applicable)

Manipulation before clinical application form is it presented for clinical application. Example:
Information associated with package, adequate storage
temperature, special requirements (e.g. need to add
solutions, cut, etc)

Identification of risks Description of the possible risks and adverse reactions


anticipated based on prior experiences and risk assessment

Clinical indication(s) of the TCTP

Minimal Follow up data required to assess


safety and efficacy

SARE monitoring and report Communication procedures with the TEs

Information on Nonclinical Studies Brief description of essays and results obtained in validation
studies and quality controls, performed before issuing the
novel TCTP for clinical application.

Version and date of the revised document

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


106
(2nd Draft – 27th March 2018 – 02(D.4.2,5.1)01(6.1,7.1,8.1))

Annex III – Template form: Methodologies for Assessing the Risks associated
to novel TCTP
(to be add in the future – See here)

Grant Agreement number: 709567 — Euro-GTP II — HP-PJ-2015


107

Potrebbero piacerti anche