19.04.2015 Views

SNBTS Research Strategy - Scottish National Blood Transfusion ...

SNBTS Research Strategy - Scottish National Blood Transfusion ...

SNBTS Research Strategy - Scottish National Blood Transfusion ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>SNBTS</strong> <strong>Research</strong>, Development &<br />

Innovation<br />

<strong>Strategy</strong> 2012-2017<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 1 of 74


Contents<br />

Page Number<br />

Introduction 3<br />

Workforce 4<br />

Financial Summary 5<br />

<br />

<br />

<br />

<br />

Publications &<br />

Grants Summary<br />

Tissue and Cellular<br />

Therapies Theme<br />

Group ~ Summary<br />

Microbiology &<br />

Components Theme<br />

Group ~ Summary<br />

Clinical <strong>Transfusion</strong><br />

Theme Group ~<br />

Summary<br />

Figure 1: Proposed Staff<br />

Structure October 2012<br />

References 9<br />

o Appendix 1 RDI Workforce Plan 2012-<br />

10<br />

2017<br />

o Appendix 2 Tissue and Cellular<br />

Therapies Theme Group<br />

Plan<br />

11<br />

o Appendix 3<br />

Microbiology &<br />

Components Theme Group<br />

Plan<br />

o Appendix 4 Clinical <strong>Transfusion</strong><br />

34<br />

Theme Group Plan<br />

o Appendix 5 Publications & Grants 42<br />

o Appendix 6 Healthcare Quality Impact<br />

Assessment Template<br />

72<br />

5<br />

6<br />

7<br />

7<br />

8<br />

23<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 2 of 74


Introduction<br />

Current <strong>SNBTS</strong> <strong>Strategy</strong> was laid out in the <strong>SNBTS</strong> Strategic Development Plan 2009-<br />

2014, approved by NSS Board in March 2009 1 . In 2011 a <strong>Strategy</strong> Refresh was initiated in<br />

order to adapt the organisation to the changing external environment including the<br />

Healthcare Quality <strong>Strategy</strong> 2010 2 , the global recession and consequent public sector fiscal<br />

retrenchment and our ageing <strong>Scottish</strong> demography. Internal drivers include retirement of a<br />

large cohort of senior and experienced staff, the requirement to find efficiency savings and<br />

the need to refocus research effort to ensure maximum quality, impact and relevance.<br />

The <strong>SNBTS</strong> <strong>Strategy</strong> Refresh 3 provides the framework for this 5 year <strong>Research</strong>,<br />

Development and Innovation plan which supports the strategic objectives in addressing the<br />

critical issues facing the organisation, particularly those around continuing threats to blood,<br />

tissue and cell safety from new and emergent infections, ensuring the continuing quality and<br />

effectiveness of clinical products and services and managing changes in demand for<br />

existing and new products due to ageing demography. The Penrose Inquiry 4 has<br />

highlighted the role of <strong>SNBTS</strong> research, development an innovation in addressing the<br />

challenges to the blood and plasma product supply posed by HIV and HCV in the 1970s<br />

and 1980s. Whilst the nature of the threats to supply, safety and quality evolve with the<br />

times, our fundamental responsibility to manage them in a timely and effective manner is a<br />

constant. Moreover, <strong>SNBTS</strong> has a broader responsibility in helping NHS Scotland to<br />

manage the forthcoming rise in chronic degenerative conditions and to support the <strong>Scottish</strong><br />

Government’s Healthcare Quality 2 and Life Sciences ambitions 5,6 .<br />

In order for <strong>SNBTS</strong> to continue to deliver high quality / impact research in support of its<br />

strategic objectives, it will focus on three research themed areas. Each will be aligned to<br />

one of the 3 <strong>SNBTS</strong> Operational Directorates, but will also interface both with the main<br />

research intensive <strong>Scottish</strong> Universities in Edinburgh, Glasgow, Aberdeen and Dundee and<br />

with partner organisations such as the <strong>National</strong> CJD Surveillance Unit and the Roslin<br />

Institute, in order to leverage a range of resources and capabilities which the organisation<br />

itself does not necessarily possess. <strong>SNBTS</strong> currently has in place formal agreements with<br />

the Universities of Aberdeen, Edinburgh and Glasgow to participate in collaborative<br />

research work in the field of transfusion medicine. Additionally, The <strong>Scottish</strong> Centre for<br />

Regenerative Medicine (SCRM) GMP cell therapy manufacturing facility is now complete<br />

and it is proposed that <strong>SNBTS</strong> will co-manage this with Roslin Cells for a period of 5 years.<br />

This will involve input from within <strong>SNBTS</strong> Tissues and Cells, Quality and RDI and this will<br />

not only facilitate NSS <strong>SNBTS</strong>’ central role in the development of cell therapy to direct<br />

clinical value to patient health but will also support the <strong>Scottish</strong> Government Life Sciences<br />

strategy in regenerative clinical and translational medicine.<br />

<strong>SNBTS</strong> will continue to work in partnership with other research-active divisions of NSS and<br />

the wider NHS Scotland, as well as with other UK and International <strong>Blood</strong> Services in order<br />

to ensure clinical relevance and efficient use of resources. Regular, quarterly joint meetings<br />

with research staff from Health Protection Scotland have been initiated to investigate areas<br />

of mutual interest, while senior staff from <strong>SNBTS</strong>, HPS and ISD are actively involved in the<br />

formulation of a <strong>Research</strong> <strong>Strategy</strong> for NSS.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 3 of 74


The proposed three new Theme Groups are:<br />

Tissue and Cellular Therapies<br />

The support of established cell and tissue therapies such as haematopoietic stem cell<br />

transplantation as well as the implementation of new cell based therapies such as<br />

pancreatic islet transplantation are both of importance.<br />

The development of a new generation of cellular therapies is a strategic objective for<br />

both the <strong>Scottish</strong> 5 and UK 7 Governments as evidenced for example by the building of<br />

the <strong>Scottish</strong> Centre for Regenerative Medicine in Edinburgh 8 and the recent<br />

announcement of funding for a Cellular Therapy Catapult Centre respectively 9 .<br />

Microbiology & Components<br />

The quality, safety and sufficiency of the blood supply is clearly the core responsibility of<br />

<strong>SNBTS</strong>. <strong>Research</strong> and development in this field is essential to ensure the organisation<br />

is prepared for the potential introduction of a vCJD screening assay on blood donation;<br />

enable rapid response to new and emergent infections and to evaluate the effectiveness<br />

and clinical safety of pathogen reduction and removal technologies.<br />

Clinical <strong>Transfusion</strong><br />

Immunological matching of blood, tissue and cells and solid organs for transplantation is<br />

key to ensuring survival of the transfused/transplanted human tissue and to manage<br />

potential adverse impacts on recipients. <strong>Research</strong> and development in this area is<br />

essential to maintain and improve the efficacy and safety of blood transfusion for<br />

patients in Scotland while also leading to the development and application of new<br />

diagnostic and therapeutic approaches.<br />

In addition there is increasing evidence to suggest that excessive transfusion particularly<br />

with aged blood can have a detrimental impact on patients. Further evaluation of this risk<br />

through both clinical and laboratory studies is essential.<br />

Workforce<br />

The strategic leadership of the <strong>Research</strong>, Development & Innovation Directorate will be<br />

performed by a Head of RDI, working with the Medical Director and with the support of the<br />

newly identified Theme Leaders to develop and implement the strategies required to<br />

achieve these organisational goals. The post of Head of RDI has been identified in the<br />

Workforce Plan as a key to this delivery and approval to fill this position has been granted.<br />

The recruitment process is underway and it is hoped the position will be filled before the end<br />

of 2012. Operational leadership will be performed by the Theme Leaders and Senior<br />

<strong>Research</strong> Scientists within each group and many of these senior staff have already been<br />

identified and are in post or will be appointed from within the existing establishment.<br />

Analysis of the probable R&D staffing complement over the next 5 years, taking into<br />

account likely retirals, options for succession by existing more junior staff, identification of<br />

critical posts and potential revenue savings has been carried out. This has considered<br />

restructuring of the staffing in each themed area, taking account of existing skills-mix and<br />

identified where gaps in the required skills and numbers are apparent. This has revealed a<br />

likely overall reduction in R&D staff numbers from 52 (2011-12) to approximately 32 (2012-<br />

13) and a recurring annual revenue saving of around 20%. This reduction and saving takes<br />

into account replacement or review of 8 posts considered to be critical for business<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 4 of 74


continuity and/or staff succession. The expected staff structure in October 2012 is shown in<br />

Figure 1 below and the summarised Workforce Plan (2012-17) is shown in Appendix 1.<br />

Financial Summary<br />

In addition to the salaries, staff related costs and goods & services budgets proposed in<br />

each of the three Theme Group sections of this report, there are costs involved in the<br />

overall management and administration of the RDI Directorate. These costs are identified<br />

below in the financial summary.<br />

<strong>Research</strong> Development & Innovation Directorate Financial Summary 2012-13<br />

Salaries Staff Related Goods & Services Total<br />

Costs<br />

RDI Central £187,726 £16,410 £96,086 £300,222<br />

Tissue & Cellular Therapies £540,086 £15,157 £67,232 £622,475<br />

Microbiology & Components £462,737 £23,409 £107,743 £593,889<br />

Clinical <strong>Transfusion</strong> £367,635 £16,019 £48,059 £431,713<br />

TOTAL £1,558,184 £70,995 £319,120 £1,948,299<br />

Publications & Grants 2008-2012<br />

Year<br />

Journal<br />

Publications<br />

Books & Book<br />

Chapters<br />

Posters<br />

&<br />

Abstracts<br />

New<br />

Grants<br />

Total Grant Income for<br />

Year<br />

(£ x 10 6 )<br />

2008/09 85 7 17 5.1<br />

2009/10 66 8 55 16 7.2<br />

2010/11 63 4 62 8 5.1<br />

2011/12 46 2 36 13 5.9<br />

Full details of publications and grants can be found in Appendix 5.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 5 of 74


Fig 1: RDI Proposed Staff Structure Oct 2012<br />

Medical Director<br />

Prof Marc Turner<br />

Head of RDI<br />

TBA<br />

Central RDI Staff<br />

<strong>Research</strong> Administrator:<br />

Isabel Ward<br />

Librarian: June MacLeod<br />

Clinical <strong>Transfusion</strong><br />

Theme Group<br />

Lead: Prof Mark<br />

Vickers<br />

Tissue & Cellular<br />

Therapies Theme<br />

Group<br />

Lead: Dr Jo Mountford<br />

Microbiology &<br />

Components Theme<br />

Group<br />

Lead: Dr Juraj Petrik<br />

Product Testing Unit<br />

Pentlands Science Park<br />

Lead: Dr Davor Fatori<br />

Staff salaries derived<br />

from <strong>SNBTS</strong><br />

Staff salaries not<br />

<strong>SNBTS</strong>-derived<br />

Aberdeen ATMU<br />

Dr Sylvia Armstrong-Fisher<br />

Dr Mike Moss<br />

Ms Sadie Henderson<br />

Ms Anne Taylor (PA)<br />

Glasgow <strong>SNBTS</strong><br />

Dr David Colligan<br />

Dr Dick Drake<br />

Dr Anne Mackie<br />

Mr Paul Hopkinson<br />

Edinburgh NSL<br />

Dr Anne Leaver<br />

University of Glasgow<br />

Dr Lamin Marenah<br />

Dr Niove Jordanides<br />

Dr Emmanuel Olivier<br />

Prof T Holyoake<br />

Ms Elaine Allan<br />

University of Edinburgh<br />

Dr L Forrester<br />

Dr P de Sousa<br />

Dr Ronnie Gallagher<br />

Ms Kay Samuel<br />

Edinburgh NSL<br />

Components<br />

Dr Ian Downing<br />

Dr Shirley MacDonald<br />

Ms Loraine McMillan<br />

Mr Alex Morrison<br />

Ms Sandie Young<br />

(Secretary)<br />

Edinburgh NSL<br />

Microbiology<br />

Mr Sandy Cleland<br />

Ms Pamela Gilhooly<br />

Dr Stuart Imlach<br />

Dr Osmany-Larralde-Diaz<br />

Ms Karen McGowan<br />

Ms Kristen Mulligan<br />

Dr Katrina Thom<br />

Clinical Services Tissues & Cells Supply Chain<br />

Clinical Services<br />

Lead: Dr Rachel Green<br />

Product Services Dept<br />

Lead: Ms Jane Pelly<br />

Microbiology Reference<br />

Unit<br />

Lead: Dr Lisa Jarvis<br />

Better <strong>Blood</strong> <strong>Transfusion</strong><br />

Group<br />

Lead: Mrs Sandra Gray<br />

vCJD Group<br />

Lead: Dr Mike Jones<br />

Other <strong>Research</strong><br />

Active Groups<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 6 of 74


Tissue and Cellular Therapies Theme Group<br />

Over the next 5 years the Cell Therapy group will build on established expertise and<br />

collaborations to concentrate our research and development studies on three blood/bone<br />

marrow derived cell populations: blood cells (including red blood cells, macrophages),<br />

vascular endothelial cells and mesenchymal stem cells. There is increasing evidence that<br />

these cell populations can be used to treat a number of serious health issues that are<br />

particularly prevalent in the <strong>Scottish</strong> population and represent significant unmet clinical<br />

need. These include cardiovascular disease, liver failure and diabetes mellitus and the<br />

continuing requirement for a secure supply of blood components for transfusion.<br />

Primary Aim: To support the development of Cellular Therapeutics and Regenerative<br />

Medicine in alignment with the <strong>Scottish</strong> Government’s Life Sciences and Healthcare Quality<br />

strategies.<br />

Scope<br />

We will make use of cells derived from adult sources (peripheral blood, bone marrow, cord<br />

blood) and from pluripotent stem cells (human embryonic SC or induced pluripotent SC);<br />

our studies will span from clarifying the applied biology of these cells to understand their<br />

differentiation and functionality, through GMP-compliant production and regulatory approval<br />

to clinical trialing for safety and efficacy. We will work closely with the <strong>SNBTS</strong> Tissues &<br />

Cells and Quality Directorates to ensure our research provides developmental support for<br />

existing <strong>SNBTS</strong> products and delivers a new generation of cell therapies. We will<br />

collaborate with the <strong>Scottish</strong> Centre for Regeneratve Medicine and international <strong>Blood</strong><br />

Services in developing our work in this field.<br />

Impact<br />

Most importantly the enhanced understanding of these cell populations and the capacity to<br />

harness their potential should lead directly to new clinical treatments for major degenerative<br />

diseases, an alternative supply of blood for transfusion and should have significant impact<br />

on the health of the <strong>Scottish</strong> population in the longer-term. Successful development of<br />

these projects and their translation into clinical practice will also result in high impact<br />

publications, leading to further external peer-reviewed funding and the generation of<br />

valuable know-how, technological advances and intellectual property that may be exploited<br />

commercially.<br />

Full details of the proposed programme of work for this Theme group can be found in<br />

Appendix 2.<br />

Microbiology & Components Theme Group<br />

Using rigorous research, development and validation techniques we expect to safeguard<br />

our preparedness for new and emerging blood borne pathogens. High quality surveillance,<br />

epidemiological and molecular data will significantly facilitate this goal. At the same time it<br />

is an imperative to continue to improve the safety of blood and tissue products for<br />

vulnerable patient groups, such as immunocompromised individuals including<br />

Haematopoietic Stem Cell and Solid Organ transplant recipients. This group of patients<br />

frequently suffer from complications caused by common pathogens immunologically<br />

controlled in the healthy population.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 7 of 74


Continued work is required on the quality and safety of the current generation of blood and<br />

tissue products.<br />

Primary Aim: To maintain a high level of microbiological safety of blood and tissue<br />

products and where possible, to improve it through the development and application of new<br />

screening technologies combined with pathogen reduction/removal measures. At the same<br />

time our aim is to continuously improve the quality and safety of blood components leading<br />

to improved clinical outcomes for patients. We will work on achieving these targets in<br />

partnership with and in support of the Donor and Manufacturing Directorates.<br />

Scope<br />

We consider a rational combination of novel multiplexing diagnostic techniques with<br />

pathogen reduction technologies the most logical way forward for the future, since no single<br />

approach is able to achieve complete microbiological blood safety on its own.<br />

Quality and safety of blood components is of critical importance for patients’ clinical<br />

outcomes. By focusing our research on reducing the causes of storage lesion and<br />

participating in international studies comparing and standardising our tests and procedures<br />

we aim at improvements in component quality, safety and logistics of their production and<br />

distribution.<br />

We will continue to collaborate with Health Protection Scotland, UK and European <strong>Blood</strong><br />

services in pursuit of these aims.<br />

Impact<br />

Achieving these goals by studying and applying new approaches and technologies, will<br />

facilitate further improvement in blood safety and quality for benefit of patients in Scotland<br />

and beyond, and maintain <strong>SNBTS</strong>’s prominent place in the forefront of <strong>Transfusion</strong><br />

Medicine.<br />

Full details of the proposed programme of work for this Theme group can be found in<br />

Appendix 3.<br />

Clinical <strong>Transfusion</strong> Theme Group<br />

Over the next five years, this group will concentrate on assessing the demand and<br />

effectiveness of blood transfusion and its potential adverse effects through epidemiological,<br />

clinical and laboratory studies. This reflects the <strong>SNBTS</strong> strategic objective to manage<br />

demand and also recent evidence implicating red cell transfusion as a cause of increased<br />

patient morbidity and mortality. Work will continue on the immunological matching of blood,<br />

tissues, cell and organs and in the development of diagnostic and therapeutic<br />

immunohaematological approaches for diseases of the blood, some of which will continue<br />

to be progressed into the commercial sector.<br />

Primary Aim: To improve the efficacy and safety of blood transfusion and tissue, cell and<br />

organ transplantation for patients in Scotland while continuing development and<br />

commercialisation of diagnostic and therapeutic approaches for diseases of the blood.<br />

Scope<br />

We will analyse the ways in which blood products are used in Scotland, paying particular<br />

attention to differences between hospitals without readily apparent explanations, as well as<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 8 of 74


oth initiate and take part in trials to assess the optimum trigger points to give transfusions.<br />

The possibility that red cell transfusions can act as harmful inducers of inflammation or<br />

thrombosis will be investigated in both clinical trials and the laboratory. We will continue to<br />

provide development and support for molecular diagnostic services as well as progress<br />

previously developed peptide based diagnostic platforms and vaccines into the commercial<br />

sector.<br />

Impact<br />

These studies will help to optimise clinical decisions concerning blood transfusion and also<br />

help to optimise the duration and conditions of blood storage, so increasing the safety of cell<br />

products provided by <strong>SNBTS</strong>. Improved immunological matching will increase the<br />

availability, quality and safety of organ and cellular transplants. Income generation from the<br />

development of new diagnostic and therapeutic materials may also result.<br />

Full details of the proposed programme of work for this Theme group can be found in<br />

Appendix 4.<br />

References<br />

1. <strong>SNBTS</strong> Strategic Development Plan. ‘Meeting the <strong>Transfusion</strong> Needs of Patients in<br />

Scotland: <strong>SNBTS</strong> <strong>Strategy</strong> 2009 – 2014. March 2009.<br />

2. NHS Scotland Quality <strong>Strategy</strong> – putting people at the heart of our NHS. May 2010.<br />

3. <strong>SNBTS</strong> <strong>Strategy</strong> Refresh April 2012- March 2017.<br />

4. The Penrose Inquiry, Preliminary Report. September 2010.<br />

5. <strong>Scottish</strong> Life Sciences <strong>Strategy</strong> 2011: Creating Wealth, Promoting Health.<br />

6. Health and Wealth in Scotland: A Statement of Intent for Innovation in Health. June<br />

2012.<br />

7. Department of Business Innovation and Skills. <strong>Strategy</strong> for UK Life Sciences.<br />

December 2011.<br />

8. http://www.lifesciencesscotland.com<br />

9. Cell Therapy Technology and Innovation Centre ‘Call For Expressions of Interest’,<br />

Technology <strong>Strategy</strong> Board, May 2011<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 9 of 74


RDI Workforce Plan 2012-2017 Appendix 1<br />

AfC Band 2012/13 2013/14 2014/15 2015/16 2016/17 Comments<br />

RDI Central 9 1 1 1 1 1 Head Of RDI, advertised June 2012. Appointment<br />

expected in last quarter of 2012.<br />

8B 0.5 0.5 0 0 0 Remainder of 1 wte transferred to Clinical <strong>Transfusion</strong><br />

5 1 1 1 1 1<br />

Sub-Total RDI Central 2.5 2.5 2.0 2.0 2.0<br />

Cell Therapy 8D 0.25 0 0 0 0 Retiral July 2012<br />

8C 0.5 0 0 0 0 Retiral Oct 2012<br />

8B 2.35 2.1 2.1 2.1 2.1 IDTransfer to M&C July; 0.7 DC, AM, RD<br />

8A 3.25 3 3 3 3 SM Transfer to M&C July<br />

7 2 2 2 2 2 KS, EA<br />

4 0.7 0.7 0.7 0.7 0.7 PH 0.7 wte<br />

Non-AfC 1.95 2.2 2.2 2.2 2.2 JM, EO & TH (0.2)<br />

(University)<br />

Sub-Total Cell Therapy 11 10 10 10 10<br />

Microbiology & Components 8C 1 1 1 1 1 JP<br />

8B 0.75 1 1 1 1 ID<br />

8A 0.75 1 1 1 1 SM<br />

7 5 5 5 5 5 AC,AM,SI,OL-D,KM(0.5)&KT(0.5)<br />

6 2 2 2 2 2 KM,LM<br />

4 2 2 2 2 2 PG, SY<br />

Sub-Total Micro & Components 11.5 12 12 12 12<br />

Clinical <strong>Transfusion</strong> 8B 3.4 3.4 3.9 3.9 2.9 RD,DC, & AM (0.3); HAL 0.5-1.0 wte;<br />

SSAF & MM 1.0 wte<br />

5 0.5 0.5 0.5 0.5 0.5 SH<br />

4 1.3 1.3 1.3 1.3 1.3 AT & PH (0.3)<br />

Non-AfC 1 1 1 1 1 LC<br />

(University)<br />

Sub-Total Clin <strong>Transfusion</strong> 6.2 6.2 6.7 6.7 6.7<br />

TOTAL 31.2 30.7 30.7 30.7 30.7<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 10 of 74


Tissue and Cellular Therapies Appendix 2<br />

Over the next 5 years the Cell Therapy group will build on established expertise and<br />

collaborations to concentrate our research and development studies on three blood/bone<br />

marrow derived cell populations: blood cells (including red blood cells, macrophages),<br />

vascular endothelial cells and mesenchymal stem cells. There is increasing evidence that<br />

these cell populations can be used to treat a number of serious health issues that are<br />

particularly prevalent in the <strong>Scottish</strong> population and present significant unmet clinical need.<br />

These include cardiovascular disease, liver failure and diabetes mellitus and the continuing<br />

requirement for a secure supply of red blood cells for transfusion.<br />

Primary Aim: To support the development of Cellular Therapeutics and Regenerative<br />

Medicine in alignment with the <strong>Scottish</strong> Government’s Life Sciences strategy.<br />

Scope<br />

We will make use of cells derived from adult sources (peripheral blood, bone marrow, cord<br />

blood) and from pluripotent stem cells (human embryonic SC or induced pluripotent SC);<br />

our studies will span from clarifying the applied biology of these cells to understand their<br />

differentiation and functionality, through GMP-compliant production and regulatory approval<br />

to clinical trialing for safety and efficacy. We will work closely with the <strong>SNBTS</strong> Tissues &<br />

Cells and Quality Directorates to ensure our research provides developmental support for<br />

existing <strong>SNBTS</strong> products and delivers a new generation of cell therapies.<br />

Impact<br />

Most importantly the enhanced understanding of these cell populations and the capacity to<br />

harness their potential will lead directly to new clinical treatments for major diseases, an<br />

alternative supply of blood for transfusion and will have significant impact on the health of<br />

the <strong>Scottish</strong> population. Successful development of these projects and their translation into<br />

clinical practice will also result in high impact publications, leading to further funding and the<br />

generation of valuable know-how, technological advances and intellectual property that may<br />

be exploited commercially.<br />

Expected Achievements [Outputs]:<br />

Project 1. To have performed first in man studies of pluripotent stem cell-derived red cells<br />

Project 2. Identified optimal cell source for the regeneration of vascular endothelium in<br />

ischemic limb disease and initiated clinical trial.<br />

Project 3. To perform a clinical trial of MSC as adjunct therapy to islet transplant.<br />

Project 4. Completion of initial clinical trials for of limbal stem cell therapy and the delivery<br />

of cytotoxic T-cell and CD133+ and/or monocyte cell therapies<br />

Benefits to <strong>SNBTS</strong> and NHS Scotland [Outcomes]:<br />

1. A cultured source of red cells that would help to manage problems of sufficiency,<br />

immune compatibility and transfusion transmitted infection as well as the potentially the<br />

adverse effects of iron loading in multi-transfused patients and from aged red cells<br />

2. A reliable GMP source of mesenchymal cells that could be used as immunomodulatory<br />

therapy for major diseases such as MS and diabetes, and haematopoietic or solid organ<br />

transplantation.<br />

3. Development of novel therapies for cardiovascular disease which represent the<br />

predominant cause of morbidity and mortality in Scotland<br />

4. Increased yield and survival of pancreatic islet isolates that will allow more patients to<br />

be transplanted and to gain longer duration of beneficial effect, helping to better manage the<br />

problem of type 1 diabetes<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 11 of 74


5. The translation of research findings to GMP compliance will allow the delivery of novel<br />

allogeneic cell therapies such as limbal stem cells and cytotoxic T-cells to clinical practice<br />

demonstrating <strong>SNBTS</strong>’s world-leading expertise in Cell Therapy development.<br />

6. The early identification of intellectual property and know-how that can be protected and<br />

exploited to generate income from new therapeutic materials, processes and diagnostic<br />

assays<br />

Project 1. <strong>Blood</strong> Cells<br />

<strong>Blood</strong> <strong>Transfusion</strong> has become a mainstay of modern medical practice. However problems persist<br />

both nationally and internationally in maintaining adequacy of supply, managing the risk of<br />

transmission of infectious agents and ensuring immune compatibility between donor and recipient.<br />

Resources to be allocated in year 1: 6.65 FTE + associated G&S/SRC = £421,420<br />

Grant funded by Wellcome Trust, SFC and SE<br />

Additional applications in progress<br />

a. Generation of GMP-grade red blood cells from pluripotent stem cells (to include application for<br />

regulatory approval for trials of PSC derived RBCs and derivation of a stable haematopoietic or<br />

erythroid progenitor population that can be used for industrial scale RBC production)<br />

Pluripotent stem cells (PSCs) have unique properties in that they can be maintained indefinitely in<br />

culture in an undifferentiated state and yet retain the ability to form all the cells and tissues within the<br />

body. They therefore offer a potentially limitless source from which to generate red cells for use in<br />

clinical transfusion. We will continue to develop methods for the production of these in vitro RBCs<br />

and perform initial in vivo safety testing and ex vivo functional assays. This work also involves the<br />

generation of haematopoietic stem cells (HSC) as an intermediate stage to RBCs. These HSC may<br />

closely resemble HSC isolated from cord blood and bone marrow and could therefore, be extremely<br />

valuable as a model to examine the maintanence and expansion of normal HSC and how they may<br />

be used more effectively for transplantation. We will specifically assess the capacity of hESCderived<br />

HSC to repopulated haematopoiesis in mice (With Dr Lesley Forrester & Prof Alexander<br />

Medvinsky; CRM, U of Ed) and investigate the effect of hypoxia on their expansion and definitive<br />

characteristics (Prof Tessa Holyoake & Dr Kamil Kranc, U of Glasgow).<br />

b. Production of induced pluripotent stem cells from adult haematopoietic cells<br />

The successful use of pluripotent cell lines for cell therapy will be dependent on immunological<br />

compatibility between the pluripotent cell line and the patient. Furthermore, the ethical concerns<br />

surrounding the use of embryonic stem cells in medical applications may limit the global uptake of<br />

such a product. A real alternative to hESC has been developed over the last 5 years, such that it is<br />

now possible to generate induced pluripotent stem cells (iPSC) that may be suitable for clinical use.<br />

We will make fully and partially reprogrammed iPSC lines at research grade in the first instance from<br />

donor tissue of defined blood group status. In the longer term we will work with Roslin Cells and the<br />

SCRM to generate GMP-grade, clinical iPSC lines. The first of these will be a type ORhD- “Universal<br />

Donor” line for RBC generation.<br />

c. Generation of monocytes from pluripotent stem cells<br />

In addition to RBC, pluripotent SC can be specifically differentiated into other blood lineages<br />

including monocytes, neutrophils and platelets. Monocytes have been tested as potential vectors for<br />

the local production of anti-inflammatory molecules that can ameliorate acute renal inflammation and<br />

offer a new therapeutic option for renal injury. The use of unmanipulated monocytes has also been<br />

proposed for liver disease (See Project 4e below). We will isolate monocytes from peripheral blood,<br />

and also generate large numbers of monocytes from PSC as a potential source of homogenous<br />

GMP-grade cells that would provide an “off the shelf” cell therapy solution for use in acutely ill<br />

patients.<br />

Project 2. Endothelial Cells<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 12 of 74


Ischaemic disease, often the result of diabetes or atherosclerosis, occurs when a patient’s blood<br />

vessels become blocked, interrupting the flow of blood to tissues and organs. Current therapies<br />

include drug therapy, bypass and angioplasty but are clearly sub-optimal since this remains a major<br />

burden to the NHS. Peripheral vascular or arterial disease has an age-adjusted prevalence of 20<br />

percent for the general population above age 70 and contributes significantly to the cost of treating<br />

CVD was estimated to cost the NHS £14.3 billion in 2006, with 20% of NHS expenditure<br />

representing direct costs of treating CVD (British Heart Foundation, 2008). Hence effective new<br />

treatments for ischaemic disease would offer therapeutic options to patients who are currently bare<br />

a high burden of morbidity and premature mortality and could make a large impact in a disease that<br />

carries a large cost to the NHS and the wider <strong>Scottish</strong> society.<br />

a. Generation of endothelial cells from pluripotent stem cells<br />

b. Isolation and expansion of endothelial progenitors from adult sources<br />

c. Generation GMP-grade endothelial cells from pluripotent stem cells<br />

The projects above will investigate the generation of vascular endothelial cells from various human<br />

SC sources (2a &b) with the dual aims of generating direct cell therapies (2c) but also elucidating<br />

mechanisms underlying this condition and identifying opportunities to develop pharmacological drug<br />

interventions to safely and locally induce angiogenesis in ischaemic tissues<br />

Resources to be allocated in year 1: 0.75 FTE + associated G&S/SRC = £51,039<br />

Addition funding from BHF, application made to MRC<br />

Project 3. Mesenchymal Cells<br />

Mesenchymal stromal cells (MSC) have been extensively studies for their capacity to regenerate<br />

skeletal tissue including bone and cartilage, however, this focus has recently shifted with the<br />

realisation that they have profound immunoregulatory capacities. These capacities are so<br />

remarkable that MSC can prevent mixed lymphocyte reactions in vitro and, in pre-clinical models,<br />

MSC can support the transplantation of allogeneic tissues. Thus, this new role for MSC as a<br />

powerful mediator of recipient immune responses is now the focus of considerable interest. The<br />

clinical utility of pluripotent stem cell-derived therapies has been questioned because of their<br />

allogeneic origin and their potential to induce immune responses and rejection in the recipient.<br />

However, immune conditioning of a recipient with MSC derived from the PSC source or even from a<br />

third party allogeneic donor, may greatly facilitate the transplantation of SC-derived tissue without<br />

rejection or immune suppressive therapy. This concept is currently being tested in Phase I/II trials to<br />

mitigate GvHD after HSC transplants. Additionally, there is growing recognition that many chronic<br />

diseases are the result of dysregulated inflammatory or immune responses, these include diabetes,<br />

multiple sclerosis, atherosclerosis, some forms of acute lung injury and liver disease. MSCs may<br />

also have the potential to re-set or regulate the immune response in these settings and could<br />

represent an important new treatment option for these intractable diseases. However, we will also<br />

continue our collaborations with academic colleagues who wish to use MSC for their established<br />

capacity to regenerate bone and cartilage. MSC can be isolated from various tissues including cord<br />

blood, we will work with the <strong>Scottish</strong> Cord <strong>Blood</strong> Bank to make use of units that have been collected<br />

under their quality systems but are not suitable for storage and transplantation.<br />

a. GMP-grade tissue collection and MSC isolation/expansion (CMT Gp)<br />

MSC are routinely isolated from bone marrow, cord blood or adipose tissue. We will make the<br />

appropriate applications to the <strong>SNBTS</strong> Committee for the Governance of Samples and also to the<br />

appropriate Local Ethics Committee to collect cord blood (rejected donations from the CB Bank),<br />

apheresis cones, adipose tissue from liposuction and waste platelet units for platelet lysate. We will<br />

establish GMP-compatible protocols in research laboratories and <strong>SNBTS</strong> “clean rooms” before<br />

transferring these to SCRM for conversion to full GMP compliant SOPs.<br />

b. Generation of MSC from pluripotent stem cells, characterisation and comparison of MSC from<br />

different sources, their capacity to modulate immune function and potential for co-transplantation<br />

We have the capacity and resource to generate these cells from many different sources at both<br />

research and GMP-grade. However, we need to look more thoroughly at the efficacy of these cells<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 13 of 74


in in vitro and in vivo assays. This work will require additional external funding and close<br />

collaboration with colleagues such as Prof S Forbes and Dr L Forrester who are already using these<br />

damage models. It will also be essential to work with the <strong>SNBTS</strong> Clinical Histocompatability &<br />

Immunogenetics group (H&I; Prof Phil Dyer) to assess the HLA-type of all tissues and cells.<br />

c. Development of MSC for co-transplantation with pancreatic tissue<br />

<strong>SNBTS</strong> in collaboration with <strong>National</strong> Service Division (NSD) has successfully introduced an Islet<br />

transplantation programme and has undertaken numerous transplantation, which have shown<br />

excellent results in the outcome for the insulin dependant diabetic recipients (n=10 in 2011), these<br />

patients have successfully recovered their glycaemic awareness and some have become insulin<br />

independent. However, the effects of the transplant can be transient and further transplantation is<br />

needed. It is well established in animal models that co-transplantation of islet cells with<br />

mesenchymal stem cells (MSCs) improves function and survival of transplanted islets. Accordingly,<br />

it is anticipated that either co-culture prior to transplantation or MSC infusion post-islet<br />

transplantation would lead to improved outcomes in islet recipients. Also, animal models have<br />

shown that the addition of MSCs to the transplantation process would provide clinical benefit with<br />

lower yields of islets, allowing good clinical outcomes with yields would be considered unsuitable for<br />

transplant and thereby increasing availability of this therapy. The GMP-compliant protocols for<br />

autologous and allogeneic MSCs developed and tested in Project 2a-c above, could be used to<br />

generate MSC as ATMPs for transplantation with an initial application for use as co-administration<br />

with islets. In addition to the established adult and hESC sources it has also been reported that<br />

MSC can be derived from the exocrine and connective tissue waste that remains after the islets<br />

have been isolated from a cadaveric pancreas for transplant. We will isolate MSC from this<br />

alternative source and investigate their use as an adjunct therapy matched to the islet graft.<br />

This work would be undertaken with Tissues & Cells Directorate, initially using <strong>SNBTS</strong> resources but<br />

with applications for external funding to be made as soon as possible.<br />

Resources to be allocated in year 1: 1.2 FTE + associated G&S/SRC = £120,108<br />

Addition input from T&C and H&I directorates<br />

Additional grant funding to be sought<br />

Project 4. Clinical Development (lead by Tissue & Cells Directorate and Product Services<br />

group with R&D support from Cell Therapy group)<br />

The majority of these projects (4b-e) are currently in translation to GMP/clinical practice and do not<br />

require immediate input from R&D staff at this time, input of scientific staff may be needed to in<br />

future to response to specific issues in development and support will be provided by CT R&D as<br />

required<br />

a. Pancreatic islets – support to TCD to enhance survival of isolated cells, develop storage<br />

protocols, identify markers on the active cell population within a graft.<br />

<strong>SNBTS</strong> Tissues & Cells, in conjunction with NHS Lothian have established a GMP compliant<br />

pancreatic islet cell isolation service for the treatment of Type I diabetic patients presenting with<br />

severe disease. The first transplant took place in early 2011 and to date there have been 16<br />

transplants in patients throughout Scotland and North-East England, with all patients demonstrating<br />

restoration of glycaemic awareness, reduced number of hypoglycaemic events, reduced insulin<br />

requirements and in some cases insulin independence. In the year 2011-12, 10 of the UK’s 19<br />

transplants were performed in Scotland. <strong>SNBTS</strong> Tissues and Cells are also able to provide a source<br />

of human pancreatic islet cells for research and development. We intend to use this material to<br />

improve the current procedure by developing quantification assays, cryopreservation and transport<br />

conditions and by correlating outcome with graft quality.<br />

Improving Islet Quality Assurance via Retrospective Analysis<br />

In collaboration with the University of Newcastle, <strong>SNBTS</strong> Tissues and Cells are currently involved in<br />

the retrospective analysis of clinical islet cell samples for quality control purposes. It is recognised<br />

internationally that the current mechanisms employed in the release of islets for transplant remain<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 14 of 74


limited, subjective and usually devoid of any functional analysis. Current methodology is restricted<br />

to the quantification of islet cell equivalents, purity and viability. <strong>SNBTS</strong> Tissues and Cells intend to<br />

collect tissue and supernatant samples from clinical isolations, which are then fixed or frozen for<br />

retrospective testing at centres throughout the UK with expertise in 3D viability analysis of beta-cells,<br />

gene and protein expression analysis and functional in vitro assays. This data will allow correlation<br />

of islet quality with clinical outcomes in transplanted patients and will ultimately aid the development<br />

of a standardised series of assays to be employed in the UK prior to clinical release of islets, thus<br />

ensuring patients are only transplanted with preparations meeting evidence-based stringent criteria<br />

that are consistent with good clinical outcomes.<br />

In addition, <strong>SNBTS</strong> Tissues and Cells plan to validate a transport box designed to control<br />

temperature and optimum physical conditions. Currently, transport of islets in the UK is highly<br />

variable with islet quality often deteriorating following extended periods in transit. Validation of such<br />

a device will ensure consistency throughout the UK and be integral to the development of an organ<br />

allocation service whereby organs are sent to the nearest centre for islet isolation, storage and<br />

subsequent transport to the recipient centre, ensuring reduced organ ischaemia, and accordingly<br />

improved isolation outcomes, and ultimately leading to increased numbers of transplants and more<br />

economical use of whole pancreata.<br />

Cryopreservation of Pancreatic Islet Cells<br />

The shortage of donor pancreata and the inability to consistently isolate large number of viable islets<br />

(average worldwide ~50%) suggests a role for the long-term storage of islets. Cryopreservation<br />

would allow indefinite storage of islet cells, leading to specific HLA matching to recipients and permit<br />

strict quality control through the ability to test all preparations for virological markers (inclusive of<br />

NAT) and microbial contamination. Although cryopreserved islets are unlikely to induce insulin<br />

independence in recipients per se, these preparations would be invaluable in ‘topping-up’ recipients<br />

via 2 nd or 3 rd transplants to sustain long-term function.<br />

Islet cryopreservation is not currently used clinically in the UK because of the lack of GMP-compliant<br />

cryopreservation equipment and storage facilities in the other UK laboratories. <strong>SNBTS</strong> Tissues and<br />

Cells have considerable experience and equipment required for controlled rate cryopreservation and<br />

considerable storage facilities required to perform GMP-translation of established protocols.<br />

Resources to be allocated in year 1: 0.55 FTE + associated G&S/SRC = £29,908<br />

Addition input from T&C directorate<br />

Additional grant funding to be sought in FY2012<br />

b. Limbal Stem Cell Study<br />

Stem cells residing in the limbal region of the eye are responsible for renewing the corneal<br />

epithelium. Limbal stem cell deficiency (LSCD) results in ocular surface disease (OSD)<br />

characterised by reduced vision or blindness, ocular irritation and visual glare. Current treatment for<br />

OSD included standard corneal transplantation and limbal tissue grafting. Both treatments have<br />

limitations with respect to tissue availability and success rates. Ex vivo expansion of limbal stem<br />

cells is therefore an alternative process for providing sufficient cells for corneal epithelium repair<br />

from small pieces of donor tissue.<br />

<strong>SNBTS</strong> is therefore involved in a study to establish a GMP process for the ex vivo expansion of<br />

limbal stem cells, from cadaveric donor tissue, on human amniotic membrane. A pilot clinical trial is<br />

being performed using this product, in collaboration with NHS Lothian, and NHS Greater Glasgow<br />

and Clyde, and will compare the outcome in 10 patients treated with the product, with 10 patients<br />

receiving an amniotic membrane control product. This is the first prospective trial to make this<br />

comparison and will provide information on whether transplantation of the ex vivo expanded corneal<br />

limbal stem cells product is feasible, efficient and safe; whether this procedure lead to improvements<br />

in vision and quality of the ocular surface; and how immunosuppression and ex vivo limbal stem cell<br />

transplantation compare with immunosuppression and amniotic membrane alone.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 15 of 74


<strong>SNBTS</strong>’s involvement in this study makes us a key player in this pilot study in that we have<br />

developed and established a suitable GMP grade product, which, following evaluation of the results<br />

of the pilot study, could lead to establishment of this product for a wider number of patients suffering<br />

from OSD. In addition, the manufacturing facility has been licensed by the MHRA as the clinical<br />

product is considered an Investigational Medicinal Product (IMP). This manufacturing licence<br />

(MA(IMP)) not only allows the facility to manufacture the LSC product, but other products considered<br />

as IMP’s. This together with the MHRA license to conduct the clinical trial has established <strong>SNBTS</strong><br />

as one of the first centres in UK to be manufacturing an Advanced Therapy Medicinal Product for<br />

clinical use.<br />

During 2012, <strong>SNBTS</strong> will also process ES derived retinal (RPE) cells for a clinical trial examining<br />

safety and efficacy in six patients with congenital retinal disease in collaboration with the US<br />

company Advanced Cell Technology (ACT).<br />

c. Cytotoxic T-cell therapy<br />

Epstein-Barr virus (EBV) is a common persistent infection controlled by cytotoxic T cells in healthy<br />

people, but can cause post transplant lymphoproliferative disease (PTLD), which is often fatal.<br />

Previously a bank of 100 cytotoxic T cell lines (CTL) has been grown from healthy blood donors and<br />

used to treat PTLD. A phase II clinical trial used these allogeneic EBV-specific showed a 52 per cent<br />

response rate at six months in PTLD patients unresponsive to all other conventional therapies. This<br />

treatment is now ready for transfer to the clinic. However, the CTL lines developed for the initial<br />

proof of principle trial were grown under good laboratory practice, but these conditions did not<br />

comply with the subsequent 2004 EU Directive on Tissues and Cells and requirements for HTA or<br />

MHRA licensure. An award from the Wellcome Trust Technology Transfer Committee has funded<br />

the creation of a new EBV specific T cell bank at <strong>SNBTS</strong> Aberdeen, a specials license has now<br />

been granted by the MHRA for the supply of these donor - derived EBV - directed cytotoxic<br />

lymphocytes. It is planned to increase the size of the bank from the current five to twenty by the end<br />

of 2013. The bank will then be maintained at a replacement level. If demand for the cells justifies<br />

further expansion, the range of HLA types available will be extended using donors from Europe and<br />

America. It is also planned to extend the utility of these cells in trials of Epstein-Barr virus<br />

associated lymphomas beyond that of the post-transplant setting. This work is undertaken by the<br />

Clinical Directorate under Prof Mark Vickers in Aberdeen with R&D support available from CT as<br />

required.<br />

d. CD133+ cells for the treatment of liver disease<br />

Liver disease is a rapidly rising cause of morbidity and mortality in the western world. Liver<br />

transplantation is currently the only curative treatment available for end stage liver disease and there<br />

is a rising demand for transplantation, which has not been matched by an increased supply of donor<br />

organs. In addition, transplantation requires lifelong immunosuppression with associated side effects<br />

including renal failure, cardiovascular complications and increased risk of malignancy. Several<br />

studies have therefore investigated the effects of stem cell therapy in liver disease in humans.<br />

However all have been small, non-randomised, and have made no attempt to formally evaluate the<br />

effect on either liver disease severity or stage of fibrosis, nor have sought to identify the mechanism<br />

by which adult stem cells exert their effect.<br />

<strong>SNBTS</strong> is therefore in collaboration with the University of Edinburgh to participate in a two centred,<br />

phase II, randomised controlled trial in liver disease. The study aims not only to evaluate two<br />

different mechanisms of cell therapy against standard conservative management but also to<br />

examine the efficacy and safety of bone marrow stem cell therapy in patients with compensated<br />

cirrhosis, and to demonstrate improvement in liver function and reduction in liver fibrosis in patients<br />

receiving these therapies compared with those receiving standard conservative management.<br />

This trial is the first randomised control trial for liver stem cell therapy, therefore <strong>SNBTS</strong>’s<br />

involvement will make a contribution to this field and establish a GMP system for preparation and<br />

cryopreservation of CD133+ selected G-CSF mobilised stem cells. Depending on the outcome of<br />

the trial, this process has the potential to be used in future for a wider number of patients suffering<br />

from liver cirrhosis.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 16 of 74


e. Monocyte/macrophage cells for liver disease<br />

This study is in collaboration with the University of Edinburgh and also aims to look at treatment of<br />

patients suffering from liver disease, but using treatment with autologous macrophages, as there is<br />

evidence from mouse models that macrophage therapy improves fibrosis, regeneration and liver<br />

function. This project is currently at the research phase with the translation to GMP planned for<br />

2012-13, with associated process validation conducted by <strong>SNBTS</strong> Cellular Therapy staff within the<br />

GMP Translational Facility of the SCRM and a clinical trial is planned for 2013. Given <strong>SNBTS</strong>’s<br />

experience in licensing of an ATMP product for use in the corneal stem cell clinical trial, we have the<br />

necessary experience and skills to progress this study to clinical trial and as such determine the<br />

outcome of this novel regenerative medicine study in humans.<br />

HEALTHCARE QUALITY IMPACT ASSESSMENT (Appendix 6)<br />

(Estimated amount of impact on patient/population: small +;medium ++ ;high +++)<br />

STUDY SAFE EFFECTIVE EFFICIENT EQUITABLE<br />

1a In vitro RBCs +++ +++ ++ ++<br />

1b iPSC from blood +++ ++ ++ ++<br />

1c In vitro monocytes/macrophages ++ ++ + +<br />

2a-c In vitro generated endothelial cells ++ +++ +++ +++<br />

3a GMP compliant MSC collection +++ +++ +++ ++<br />

3b In vitro generation of MSC from ++ +++ +++ ++<br />

optimal source<br />

3c MSC from pancreatic tissue +++ +++ +++ ++<br />

4a Pancreatic islet optimisation +++ +++ +++ +++<br />

4b Limbal Stem cell Study ++ +++ ++ +<br />

4c Cytotoxic T-cell therapy ++ +++ + ++<br />

4d CD133+ cells for liver disease ++ +++ + +<br />

4e Monocytes/macrophages for liver<br />

disease<br />

++ ++ + ++<br />

None of these projects is expected to impact on timeliness or person centeredness.<br />

Total resources allocated FY 2012-13<br />

Project Staff Cost G&S SRC TOTAL<br />

1. <strong>Blood</strong> cells £362,934 £49,219 £9,267 £421,420<br />

2. Endothelial cells £43,680 £6,082 £1,277 £51,039<br />

3. Mesenchymal stem cells £106,905 £9,571 £3,632 £120,108<br />

4. Pancreatic islets and<br />

£26,567 £2,360 £981 £29,908<br />

clinical support<br />

TOTAL £540,086 £67,232 £15,157 £622,475<br />

5 yr Project Objectives<br />

Project 1. To have performed first in man studies of pluripotent stem cell-derived red cells<br />

Project 2. Identified optimal cell source for the regeneration of vascular endothelium in ischemic<br />

limb disease and initiated clinical trial.<br />

Project 3. To perform a clinical trial of MSC as adjunct therapy to islet transplant.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 17 of 74


Project 4. Completion of initial clinical trials for of limbal stem cell therapy and the delivery of<br />

cytotoxic T-cell and CD133+ and/or monocyte cell therapies to clinical practice<br />

Project Milestones (TBC = To be completed by)<br />

Project Objective Yr 1<br />

04/12-03/13<br />

1<br />

Initiate FIM Complete initial<br />

PSC RBC studies of WT grant<br />

pluripotent stem<br />

cell derived<br />

RBCs<br />

Yr 2<br />

04/13-03/14<br />

Initial detailed<br />

discussions<br />

with<br />

EMA/MHRA<br />

Milestones<br />

Yr 3<br />

04/14-03/15<br />

Detailed<br />

discussions<br />

with<br />

EMA/MHRA to<br />

finalise<br />

requirements<br />

for application<br />

Yr 4<br />

04/15-03/16<br />

Plan FIM safety<br />

trial in healthy<br />

volunteers and<br />

phase I in<br />

thalassaemia<br />

patients<br />

Yr 5<br />

04/16-03/17<br />

Initiate FIM trial<br />

in healthy<br />

volunteers<br />

TBC Dec 12<br />

Apply for short<br />

term extension<br />

of WT grant to<br />

Apr 13<br />

TBC Mar 14<br />

TBC Mar 15<br />

TBC Sept 15<br />

Compile<br />

regulatory<br />

application and<br />

discuss with<br />

EMA/MHRA<br />

TBC Mar 17<br />

Finalise<br />

regulatory<br />

application<br />

TBC July 12<br />

Apply for 2nd<br />

phase WT STA<br />

award to run<br />

4/13-3/17<br />

Initiate scale-up<br />

studies<br />

incorporating<br />

new partners if<br />

necessary<br />

TBC Mar 16<br />

TBC Mar 17<br />

TBC Dec 12<br />

Continue basic<br />

science studies<br />

to enhance<br />

enucleation &<br />

maturation<br />

Rolling<br />

Continue basic<br />

science studies<br />

to enhance<br />

enucleation &<br />

maturation<br />

Finalise<br />

protocol for<br />

production of<br />

enucleated<br />

erythrocytes<br />

In vitro & in vivo<br />

testing of<br />

enucleated<br />

RBCs<br />

Rolling<br />

Produce 1011<br />

nucleated<br />

erythroid cells<br />

under GMP at<br />

Roslin Cells<br />

Rolling<br />

GMP<br />

conversion of<br />

protocols<br />

TBC Mar 15<br />

TBC Mar 16<br />

Finalise GMP<br />

grade protocol<br />

for production<br />

of enucleated<br />

erythrocytes<br />

Produce 1013<br />

GMP grade<br />

enucleated<br />

erythrocytes<br />

1<br />

PSC HSC<br />

Mar 2017<br />

Identify the<br />

repopulating<br />

capacity of PSC<br />

derived HSC<br />

TBC Mar 13<br />

Generate first<br />

reprogrammed<br />

cell lines from<br />

PB MNCs<br />

TBC Mar 13<br />

Application for<br />

funding to LLR<br />

Rolling<br />

Convert<br />

research<br />

protocol for<br />

reprogramming<br />

to GMP<br />

TBC Mar 14<br />

Application to<br />

MRC Regen<br />

Med Platform<br />

for <strong>Blood</strong><br />

Program<br />

Produce GMP<br />

grade<br />

reprogrammed<br />

cells<br />

(with Roslin<br />

Cells)<br />

TBC Mar 15<br />

TBC Sept 15<br />

Differentiate<br />

RBC from GMP<br />

reprogrammed<br />

cells<br />

TBC Mar 16<br />

TBC Jul 16<br />

Generate first<br />

reprogrammed<br />

cell lines from<br />

PB MNCs<br />

TBC Mar 13<br />

Mar 2016<br />

TBC Mar 13<br />

Identification of<br />

sub-populations<br />

of PSC derived<br />

HSC/HPC over<br />

timecourse<br />

TBC Mar 14<br />

Complete in<br />

vitro<br />

assessment of<br />

HSC/HPC on<br />

stroma<br />

Transfer<br />

findings to other<br />

blood projects<br />

where<br />

applicable<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 18 of 74


TBC Mar 13 TBC Mar 14 Rolling<br />

Initiate in vivo<br />

transplant<br />

assays<br />

In vivo<br />

transplant<br />

assays<br />

Complete long<br />

term in vivo<br />

repopulation<br />

assays<br />

Assessment and<br />

publication of<br />

the effect of<br />

hypoxia on the<br />

expansion and<br />

maintenance of<br />

HSC from PSC<br />

and adult<br />

sources<br />

Mar 2015<br />

Transfer of staff<br />

to project<br />

TBC Oct 12<br />

Complete staff<br />

training in<br />

specific<br />

techniques<br />

TBC Mar 14<br />

Complete<br />

optimisation of<br />

haemopoietic<br />

assays under<br />

normoxia and<br />

various hypoxic<br />

conditions<br />

TBC Apr 13<br />

In vitro assays)<br />

for epansion of<br />

HSCs/HPCs<br />

under hypoxic<br />

conditions<br />

Rolling<br />

In vivo assays<br />

of HSCs/HPCs<br />

expanded<br />

under hypoxic<br />

conditions<br />

TBC Mar 15<br />

Complete<br />

titration of PHD<br />

inhibitors in<br />

HSCs/HPCs.<br />

Initial<br />

characterisation<br />

of HSC/HPC<br />

functions<br />

following PHD<br />

inhibition<br />

TBC Mar 16<br />

TBC Mar 13<br />

TBC Mar 14<br />

TBC Mar 15<br />

Generation of<br />

HIF and Cited2<br />

knockdown<br />

HSCs and<br />

generation of<br />

RNA for RNA<br />

sequencing<br />

TBC Mar 15<br />

Assess<br />

progress and<br />

re-plan<br />

accordingly<br />

1 PSC<br />

Monocyte<br />

Identify whether<br />

the generation of<br />

monocytes from<br />

pluripotent stem<br />

cells is a viable<br />

clinical option<br />

Differentiation<br />

of monocytes<br />

from hESC<br />

Assess protocol<br />

for GMP<br />

translation<br />

TBC Mar 15<br />

Close out if not<br />

clinically<br />

applicable<br />

Mar 2014<br />

TBC Mar 13<br />

Identification of<br />

specific<br />

monocytes &<br />

macrophage<br />

sub groups that<br />

can be<br />

generated from<br />

hESC<br />

TBC Mar 14<br />

Apply for further<br />

funding<br />

TBC Mar 15<br />

Close out if<br />

funding not<br />

sucessful<br />

2 Endoth<br />

Cells<br />

Identify optimal<br />

cell source for<br />

the regeneration<br />

TBC Mar 13<br />

Optimisation of<br />

research SOP<br />

to produce<br />

TBC Mar 14<br />

TBC May 15<br />

Complete in<br />

vivo testing of<br />

hESC derived<br />

Secure further<br />

funding or close<br />

out<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 19 of 74


of vascular<br />

endothelium in<br />

ischemic limb<br />

disease and<br />

initiated clinical<br />

trial<br />

endothelial cells<br />

from hESC<br />

endothelial cells<br />

Mar 2017<br />

TBC Mar 13<br />

Application to<br />

BHF MBH<br />

Initiative for a<br />

Centre for<br />

Vascular<br />

regeneration.<br />

TBC Mar 15 TBC Dec 15<br />

Finalise<br />

protocol for<br />

GMP isolation<br />

and expansion<br />

of pericytes<br />

from adipose<br />

tissue (SCRM)<br />

Grant<br />

application to<br />

fund<br />

collaboration<br />

with Roslin<br />

Cells<br />

EPSRC-CASE<br />

PhD<br />

studentship to<br />

start (11/12)<br />

TBC Apr 13<br />

Conversion of<br />

research SOPs<br />

to GMP<br />

TBC Mar 15<br />

GMP<br />

production of<br />

10 9 endothelial<br />

cells from hESC<br />

Secure further<br />

funding or close<br />

out<br />

3 MSC Initiate clinical<br />

trial of GMP<br />

MSCs for<br />

immunomodulati<br />

on<br />

Sept 2015<br />

TBC Jul 12<br />

Establish ethics<br />

and SOPs for<br />

tissue collection<br />

at GMP,<br />

including<br />

platelet lysate,<br />

peripheral<br />

blood/buffy<br />

coats, cord<br />

blood<br />

TBC Mar 13<br />

Finalise<br />

research grade<br />

SOP for<br />

isolation and<br />

expansion of<br />

adult MSC<br />

TBC Mar 14<br />

Apply for<br />

external funding<br />

Initial<br />

discussion with<br />

EMA/MHRA<br />

TBC Mar 14<br />

Translation<br />

research SOP<br />

to GMP<br />

TBC Mar 15<br />

TBC Dec 15<br />

TBC Mar 13<br />

TBC Sept 13<br />

Routine<br />

isolation and<br />

expansion of<br />

MSC from adult<br />

sources at GMP<br />

(SCRM) 10 8<br />

cells per<br />

sample<br />

TBC Dec 13<br />

Define potency<br />

assays<br />

TBC Jan 14<br />

Regulatory<br />

application to<br />

EMA/MHRA<br />

Initiate trial<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 20 of 74


3<br />

PSC MSC<br />

GMP production<br />

of PSC derived<br />

MSC<br />

Full<br />

phenotypical<br />

characterisation<br />

of MSC from<br />

hESC<br />

compared to<br />

adult<br />

TBC Mar 14 TBC Sep 14<br />

Complete Establish GMP<br />

comparison of SOPs for<br />

cytokine profile production of<br />

of ESC-MSC hESC-MSC<br />

and adult MSC<br />

TBC Mar 13<br />

TBC Mar 14<br />

Establish<br />

appropriate in<br />

vivo models<br />

with<br />

collaborators at<br />

UofG and CRM<br />

Edinb<br />

TBC Mar 15<br />

Apply for<br />

external funding<br />

3/4<br />

Pancreas<br />

MSC<br />

Mar 2015<br />

Initiate clinical<br />

trial of MSC as<br />

adjunct therapy<br />

to islet<br />

transplant for<br />

regulatory<br />

approval<br />

Isolate MSC<br />

from exocrine<br />

pancreatic<br />

tissue<br />

TBC Mar 13<br />

TBC Mar 14<br />

Apply for<br />

external funding<br />

TBC Apr 13<br />

Complete<br />

characterisation<br />

of MSC from<br />

pancreas and<br />

comparision to<br />

hESC-MSC and<br />

adult MSC in<br />

MLR, cytokines<br />

assays<br />

TBC Mar 15<br />

In vitro and in<br />

vivo pre-clinical<br />

testing<br />

Compile<br />

regulatory<br />

application<br />

EMA/MHRA<br />

for<br />

Initiate trial<br />

4<br />

Clinical<br />

Devt<br />

Cornea<br />

4<br />

Clinical<br />

Devt<br />

Retina<br />

4<br />

Clinical<br />

Devt<br />

Mar 2016<br />

Complete initial<br />

clinical trial of<br />

limbal stem cell<br />

therapy<br />

Mar 2015<br />

Complete<br />

clinical trial of<br />

RPE cells for<br />

Stargadt’s<br />

disease, with<br />

ACT<br />

Mar 2016<br />

Clinical delivery<br />

of cytotoxic T-<br />

cell therapy<br />

Initiate<br />

trial<br />

clinical<br />

TBC Mar 13<br />

Expansion of<br />

cell bank to 25<br />

deposits<br />

TBC Mar 14<br />

Plan expansion<br />

of trial beyond<br />

post-transplant<br />

(PTLPD) setting<br />

TBC Mar 15<br />

Complete<br />

recruitment<br />

TBC Mar 15<br />

Complete<br />

recruitment<br />

TBC Mar 15<br />

Initiate trial<br />

TBC Mar 16<br />

Complete<br />

recruitment<br />

CTL<br />

4<br />

Clinical<br />

Devt<br />

Pancreas<br />

Mar 2017<br />

Delivery of an<br />

improved<br />

pancreatic islet<br />

graft therapy<br />

TBC Mar 13<br />

Complete initial<br />

viability and cell<br />

marker<br />

characterisation<br />

studies of<br />

pancreatic islets<br />

with T&C group<br />

TBC Mar 13<br />

TBC Jun13<br />

Apply for<br />

external funding<br />

TBC Dec 13<br />

TBC Mar 15<br />

TBC Dec 16<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 21 of 74


Complete<br />

retrospective<br />

analysis of graft<br />

characterisation<br />

vs clinical<br />

outcome<br />

Direction<br />

dependent<br />

Yr1-2<br />

on<br />

4<br />

Clinical<br />

Devt<br />

CD133<br />

4<br />

Clinical<br />

Devt<br />

Monocyte<br />

Mar 2017<br />

Complete initial<br />

clinical trials for<br />

CD133+ cell<br />

therapy for liver<br />

disease<br />

Mar 2017<br />

Complete initial<br />

clinical trials for<br />

monocyte cell<br />

therapy for liver<br />

disease<br />

Mar 2017<br />

Initiate trial in<br />

Edinburgh<br />

TBC Mar 13<br />

Application for<br />

trial approval<br />

and funding<br />

TBC Mar 13<br />

TBC Mar 14<br />

Initiate trial in<br />

Edinburgh<br />

TBC Mar 13<br />

Complete<br />

recruitment<br />

TBC Mar 16<br />

Complete<br />

recruitment<br />

TBC Mar 16<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 22 of 74


Microbiology & Components Appendix 3<br />

Theme Group:<br />

Microbiology and Components (MC) R&D<br />

Our mission is to maintain high level of microbiological safety of blood and tissue supply,<br />

and where possible, to improve it through the development and application of new<br />

diagnostic technologies, combined with pathogen reduction/removal measures. At the<br />

same time our aim is to continuously improve the quality and safety of blood components<br />

leading to improved clinical outcomes for patients. We work on achieving these targets in<br />

partnership with and in support of the Supply Chain.<br />

Summary of Support Proposed:<br />

Salary: £462,737 13 staff (12FTE)<br />

SRC: £23,409<br />

G&S: £107,743<br />

Expected Achievements [Outputs]:<br />

1. Evaluation of the availability of commercial or in-house assays for emerging blood borne<br />

pathogens.<br />

2. Evaluation of the proportion of common viruses harmful to immunocompromised<br />

recipients, originating in blood and blood products<br />

3. Determination of the hepatitis E virus seroprevalence and RNA frequency in <strong>Scottish</strong><br />

blood donors and its threat to blood supply<br />

4. Identification of synthetic polymer ligands for difficult-to-inactivate viruses, for their<br />

potential removal<br />

5. Evaluation of the suitability of new diagnostic platforms for blood testing and safety<br />

margin increase<br />

6. Evaluation of the effects of new platelet and red cell additive solutions on storage and<br />

quality of blood components<br />

7. Investigation of beneficiary effect of a kinase –inhibitor on platelet storage lesion<br />

8. Determination of key platelet activation pathways through a comparison of the activation<br />

by natural and synthetic activators<br />

9. Input into validations (with Supply Chain) for new microbiology assays and components<br />

safety and quality improvements<br />

10. Validation of vCJD confirmatory assay<br />

11. Determination of Borrelia burgdorferi seroprevalence in <strong>Scottish</strong> blood donors<br />

Benefits to <strong>SNBTS</strong> and NHS Scotland [Outcomes]:<br />

1. Improvements of microbiological safety of blood supply through up-to-date diagnostics<br />

2. Improvements of microbiological safety of <strong>Scottish</strong> blood supply through new pathogen<br />

reduction/removal materials and techniques<br />

3. Safety and quality improvements in storage and provision of blood components<br />

4. Building on a successful income generation through IP and technology sale (eg<br />

microarray blood grouping), by developing new diagnostics and pathogen reduction<br />

materials and techniques<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 23 of 74


Proposed projects:<br />

MC1: Epidemiology, pathogenesis and diagnostics of blood borne pathogens<br />

1a: Impact of new and emerging pathogens on <strong>Scottish</strong> <strong>Blood</strong> and Tissue Supply<br />

Indigeneous cases of West Nile virus, Chikungunya, Dengue, Usutu and Chagas disease<br />

have all been described in some European countries and epidemiologists do not rule out<br />

the possibility of certain mosquito-borne pathogens reaching north European countries. But<br />

even substantial spread to traditional southern tourist destinations (Spain, France, Italy and<br />

Portugal) may have potentially highly damaging effect on UK blood supply. We have to<br />

have at our disposal reliable validated assays - commercial or developed in-house - suitable<br />

to deal with the potential spread of these infections. Together with MRU we are evaluating<br />

in house and commercial WNV assays to be selected as a confirmatory assay after<br />

introduction of WNV screening in Scotland in 2013. Assay development for other abovementioned<br />

targets will depend on SACTTI risk assessments for UK and availability of<br />

commercial assays. Hepatitis E is a zoonosis in developed countries and we are assessing<br />

the threat to <strong>Scottish</strong> blood supply in project 1bII (below).<br />

Within NSS, we are looking for ways to complement <strong>SNBTS</strong> laboratory research with<br />

surveillance techniques used by Health Protection Scotland (HPS) for epidemiological<br />

studies. Since we face similar problems with other UK blood services, we aim at<br />

coordination of our approaches with NHS BT, Welsh and Irish BT.<br />

1b: Impact of common viruses potentially transmitted through blood components on<br />

immunosuppressed recipients.<br />

1bI) Epidemiology and impact of human papillomaviruses, polyomaviruses, herperviruses 6,<br />

7, 8 and parvovirus B19 on immunosuppressed individuals<br />

CSO grant application was not funded. A large part of this application has now been<br />

incorporated into the NIHR/MRC EME program application coordinated in Cambridge<br />

(submitted June 2012), using new card PCR diagnostics. Another part will be used in an<br />

FP7 application also coordinated in Cambridge (1st stage of the application process<br />

October 2012).<br />

Immunosuppression commonly leads to reactivation and increased susceptibility to<br />

infection. Majority of viral replication are represented by reactivated viruses, in a proportion<br />

a superinfection or primary infection would originate from transplanted organ/tissue and<br />

transfused blood products. This proportion is currently unknown in many cases, but needs<br />

to be established as a part of duty of care, so that any measures necessary to<br />

reduce/eliminate these transmissions can be developed and implemented.<br />

1bII) Epidemiology and identification of potential transmission routes of autochthonous<br />

Hepatitis E virus (HEV) in Scotland and clinical relevance of HEV in chronic liver disease.<br />

ETM 32 is a collaboration between Glasgow Caledonian University, <strong>SNBTS</strong>, Norwich<br />

University Hospital Glasgow Royal Infirmary, Edinburgh Royal Infirmary Royal Cornwall<br />

Hospital Truro and Health Protection Scotland.<br />

This is CSO-funded project (ETM 32).<br />

Unlike HepE in endemic regions, HepE acquired in developed countries appears to be<br />

caused by genotype 3 (genotype 4 in China and Japan). Data also suggests that<br />

subclinical and/or unrecognised HepE infection is common and there is evidence to suggest<br />

that zoonotic transmission between swine (possibly shellfish) and humans may be<br />

responsible.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 24 of 74


Transmission of HEV to humans by eating infected meat has been demonstrated. In<br />

addition, HEV transmission via blood components has been reported. Chronic HEV<br />

infection was reported in immunosuppressed individuals.<br />

In order to assess the risk posed by HEV infection and facilitate suitable preventive<br />

measures, further epidemiological investigations are essential. Studies to date have been<br />

conducted in England (~10 - 13 % seroprevalence) with an absence of corresponding data<br />

for Scotland. So far we have carried out ELISA antibody prevalence study on majority of<br />

planned samples. Prevalence in <strong>Scottish</strong> donors determined to date is substantially lower<br />

than in England (~4.8 % vs 10-13%), closer to figures determined for New Zealand. PCR<br />

study to detect any active infection in blood donors, using plasma minipools is ongoing.<br />

1c: Effect of mutants on routine assays<br />

A separate issue is how efficient the routine current assays are in detection of viral<br />

variants/mutants. Some missed samples by certain HIV PCR assays have been highlighted<br />

recently, and similar situation have been described for hepatitis B surface antigen (HBsAg)<br />

assays. We have investigated this problem by cloning, sequencing and expression of the<br />

HBsAg from identified achived donations and found a new mutant in the process. We plan<br />

to expand on these studies in collaboration with NMRU, pending external funding. It is<br />

important to establish the molecular background behind the detectability of mutants by<br />

various assays. We have been in touch with Abbott where they have an established<br />

research program for characterisation of mutants.<br />

MC2: New diagnostic and pathogen reduction technologies for safer blood provision<br />

The safety of transfused blood and blood products in relation to blood borne pathogens is<br />

achieved through a combination of measures: donor selection, testing of donated blood and<br />

use of pathogen reduction/inactivation technologies (PRT).<br />

PRT represents a viable alternative to bacterial testing, viable replacement for gamma<br />

irradiation to avoid GVHD; it is generally efficient for enveloped viruses; could potentially<br />

reduce unknown pathogens and pathogen levels during window period (even if not<br />

eliminated completely). PRT however suffers from a lower efficiency of<br />

reduction/inactivation for some non-enveloped viruses, bacterial spores and does not<br />

inactivate protein-only infectious agents such as vCJD. It introduces extra steps during<br />

processing; any PRT leads to certain loss of blood component efficacy.<br />

In the situation where testing cannot guarantee 100 % detection and PRT cannot guarantee<br />

100 % inactivation, the most logical approach would be a rational combination of screening<br />

and PRT. Even if PRT could improve to a level it could guarantee a safe blood supply, it<br />

needs to be complemented by the efficient multiplexing testing platform acting as a Quality<br />

assurance tool. Microarrays, new nucleic acid amplification technologies and next<br />

generation sequencing (NGS) can provide such multiplexing platforms.<br />

As a rule, these technologies are being developed for much wider applications than blood<br />

testing, which is a niche market. It means that the task of monitoring and evaluating these<br />

new technologies for their suitability and potential adoption as next generation blood testing<br />

platforms depends on blood services’ R&D units.<br />

MC2a: Synthetic polymer ligands for removal of difficult-to inactivate non-enveloped<br />

viruses.<br />

Seed funding (12 months) was awarded by the Foundation Alliance Biosecure (France).<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 25 of 74


We are planning to apply for an extension of this study and/or a joint application with<br />

Professor Mark Bradley, School of Chemistry, University of Edinburgh for Proof of Concept<br />

funding (or similar) for this project and MC3b(also exploiting synthetic polymer libraries)<br />

Based on current performance of PRT, in order to improve blood safety we still need<br />

alternative and/or complementary approaches. One of them could be a removal of difficultto-inactivate<br />

non-enveloped viruses using specific reagents. Such approach is being<br />

evaluated for prions (prion filters) which due to their chemical nature cannot be inactivated<br />

by current PRT which is based on nucleic acid inactivation. Synthetic polymers are more<br />

suitable for large scale application than more expensive, more difficult to prepare and more<br />

labile antibody-based reagents. Availability of a virus removal option would further improve<br />

blood safety.<br />

MC2b: Novel diagnostic approaches and technologies: microarrays, biochips,<br />

nucleic acid amplification technologies (NAT) and next generation sequencing (NGS)<br />

2-year collaborative project on the development of viral microarray for mandatory targets<br />

was supported until November 2011 by Alba Bioscience.<br />

For 2012 we have been invited to join the application for FP7 European funding, lead by the<br />

University of Lyon and Etablissement Francais du Sang (EFS) Montpellier.<br />

As mentioned above microarrays, new nucleic acid amplification technologies and next<br />

generation sequencing (NGS) can provide multiplexing platforms. Monitoring, development<br />

and evaluation of novel diagnostic technologies which are suitable or can be adopted for the<br />

highly specialised and regulated field of blood testing is important for future blood bank<br />

operations and improved safety of blood supply. Future development will determine if<br />

diagnostics or PRT will be dominant in blood safety. Nevertheless, any scenario will<br />

depend on a rational combination of best components of the two approaches. In diagnostics<br />

it means the development and selection of a cost-efficient and reliable multiplexing platform.<br />

Since the commercial launch of the massively parallel pyrosequencing platform, the last 5<br />

years have been dominated by the relentless progress of high throughput genomic analysis,<br />

termed next generation sequencing (Metzker, 2005). What really positioned NGS to be<br />

considered for diagnostic applications was continuous reduction in the cost of runs,<br />

contributed to also by the NHGRI’s Revolutionary Genome Sequencing Technologies<br />

program, aimed at the sequencing of a human genome for $1000 or less.<br />

In transfusion medicine, targeted sequencing could identify a variety of relevant blood-borne<br />

viruses, bacteria and parasites, present in individual samples, including transplant recipients<br />

with potentially a number of replicating pathogens due to the immunosuppression.<br />

Application of NGS to genotyping of complex blood group systems is already being<br />

addressed (Stabentheiner et al, 2011). It can also be expected that NGS analysis of newly<br />

produced cell lines and differentiated cells resulting from stem cell technology will become a<br />

requirement. The availability of lower throughput, more economical benchtop NGS<br />

instrument makes their use realistic and future <strong>SNBTS</strong> R&D would undoubtedly be<br />

accelerated by the system availability. Joint capital bid of Microbiology and Components<br />

theme group with Tissue and Cellular Therapy theme group (Dr. J. Mountford) is proposed.<br />

A lease for the technology evaluation (but not for stem cell-derived lines quality control)<br />

would be a less favourable option which could be pursued if capital funding was<br />

unsuccessful.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 26 of 74


MC3: Improvements in quality and safety of blood components<br />

The main purpose of studies included in this project is included in the title. We are trying<br />

continuously to find ways to reduce the storage lesion, while positively effecting the storage<br />

period, and to increase the microbiological safety of components. Some of the studies are<br />

initiated by BEST-collaborative, some performed in collaboration with manufacturers<br />

(egPAS5).<br />

3a: Effect of preparation, storage conditions, pathogen reduction on quality of blood<br />

components and their metabolic state.<br />

Rationale and impact<br />

All three studies (A-C) in this project relate to the quality of platelets investigating various<br />

aspects of storage lesion and mechanisms of platelet activation, which are not completely<br />

understood. They also relate to a safety aspect, especially PAS 5 study on replacing 99%<br />

of plasma with new platelet additive solution and reducing potential for vCJD transmission<br />

and allergic transfusion reactions. These studies usually produce pilot data for any potential<br />

subsequent validation studies (see MC4: Validations). Some of these studies are initiated<br />

by BEST-collaborative group (<strong>SNBTS</strong> is a member of the group) allowing for participation<br />

and standardisation of various methods. Studies in this project will be updated/initiated<br />

accordingly.<br />

3aI) Platelet additive solution (PAS) 5 (Collaboration with and reagent support from<br />

Fenwall)<br />

Study of >99% plasma replacement in apheresis platelet donations with PAS5, a new<br />

platelet additive solution, in order to reduce the risk of vCJD transmission and to prevent<br />

allergic transfusion reactions.<br />

Platelets for transfusion are collected in 100% donor plasma. This plasma can cause<br />

transfusion reactions in some patients. It has also been shown that secondary transmission<br />

of vCJD prions can occur by transfusion of blood components including plasma. In addition,<br />

some pathogen inactivation technologies seem to work better in PAS. Therefore, being<br />

able to remove almost 100% of the plasma would be beneficial. Two different apheresis<br />

machines, Amicus and Trima, will be tested and, therefore, 20 units in total will be required.<br />

A range of tests looking at platelet morphology, metabolism, activation, lysis and reflection<br />

of in vitro function will be performed. Testing will be carried out until day 7, evaluating the<br />

potential for shelf life extension.<br />

3aII) p38 MAPK inhibitor<br />

During storage, platelets undergo various biochemical and structural changes collectively<br />

known as the platelet storage lesion. Based on new data on platelet metabolism and<br />

pathways, we intend to determine if use of specific inhibitory molecules could reduce the<br />

platelet storage lesion.<br />

It has been demonstrated recently that p38 MAPK plays a role in regulation of some of<br />

these processes. p38 MAPK inhibitor resulted in less activation and less glycocalicin<br />

production, but DMSO used as a solvent appears to counteract any benefits. We have<br />

decided to extend the initial studies using alternative solvent(s) for inhibitor (SB203580) and<br />

include soluble P-selectin measurement in order to determine if low levels of CD62P (Pselectin)<br />

are due to low activation or to cleavage.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 27 of 74


3aIII) Platelet activation: molecular analysis (Collaboration with the School of Chemistry<br />

UoE)<br />

Mechanisms of platelet activation are not fully understood. Using a comparative model of<br />

natural and synthetic polymer platelet activators which we identified earlier (Hansen et al,<br />

2011, Biomaterials, 32, 7034), we aim to analyse, at a molecular level, if natural and<br />

synthetic activators activate the same or different pathways. We intend to use the obtained<br />

data to identify specific metabolic steps and/or inhibitors which could be targeted to reduce<br />

the storage lesion.<br />

3b: Use of synthetic polymer ligands for separation and evaluation of fresh and aged<br />

blood.<br />

Rationale and impact<br />

There is a longstanding debate on potential negative effect of aged blood on clinical<br />

outcomes. There are numerous papers in support and those opposing this concept. We do<br />

not have many reliable markers for aging erythrocytes and the subpopulations are usually<br />

separated by centrifugation, based on changing erythrocyte shape/size/density. An<br />

alternative can be provided by separation of populations of different age using specific<br />

synthetic polymer ligands. This would facilitate studies leading to clarification of this issue,<br />

extremely important for clinical practice.<br />

There is a direct link to the Clinical <strong>Transfusion</strong> new grant-funded project investigating how<br />

transfused red cells might cause immune activation and dysfunction. Any new set of<br />

markers and/or separation method could be helpful within <strong>Blood</strong>Pharma project on in vitro<br />

erythrocyte production using stem cell technology (Cell Therapy).<br />

Technically, this project will use synthetic polymer libraries prepared by the School of<br />

Chemistry, UoE, similarly to project MC2a searching for ligands for specific virus removal.<br />

MC4: Validation studies for Components and Microbiology: Interface between<br />

Supply Chain and R&D<br />

Rationale and Impact:<br />

It has been recognised that there is a need for a Validation laboratory, representing a link<br />

between R&D and Supply chain for various validation studies and which is crucial for a<br />

successful transition of methods and procedures from the laboratory to routine use.<br />

4a: Overnight (O/N) hold<br />

This validation may impact substantailly on the delivery of blood and blood components<br />

from manufacturing (P&T) sites to clinical banks, improving the logistics of the process.<br />

Summary:<br />

Maco Pharma B & T bags.<br />

Short hold = Whole blood held O/N and processed 14 – 16h post donation.<br />

Long hold = Whole blood held O/N and processed 22 – 24h post donation.<br />

- 20 short hold units and 20 long hold units will be tested on day 35;<br />

- 20 short hold and 20 long hold units, irradiated on day 14, will be tested on days 15 and<br />

28.<br />

4b: Platelet Pathogen Reduction (lead by Supply Chain)<br />

Pathogen reduction technologies are in use in over 80 centres in more than 15 countries.<br />

The most frequently quoted reasons for introduction were problems with bacterial testing,<br />

replacement of gamma irradiation, inactivation of emerging pathogens and extension of<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 28 of 74


platelet shelf life to 7 days (see also MC2). In Scotland 7 day platelets would help to reduce<br />

the wastage significantly, especially in more remote northern regions.<br />

The validation protocol will be prepared in co-operation with the Supply Chain and selected<br />

provider of PRT, a health economic evaluation will also be carried out and a pilot study will<br />

be run.<br />

4c: Additive solutions (candidate solutions : PAS5, SOLX)<br />

PAS5 is currently a subject of initial evaluation study (see MC3a). The preliminary data is<br />

encouraging. If the final study outcome is equally positive PAS5 will be proposed for a<br />

validation study.<br />

SOLX: At the most recent BEST-collaborative meeting, a promising data was presented on<br />

the extension of erythrocyte storage after various periods of initial rt hold. We plan to do an<br />

initial small scale study if it can be agreed with the manufacturer. SOLX validation would be<br />

a logical extension of 4a: Overnight hold.<br />

MC 5: Non-Viral Pathogens<br />

A) Development of PMCA as a variant CJD confirmatory assay.<br />

Detection of the abnormal form of the prion protein (PrP Sc ): WO 2011/138578.<br />

PMCA (Protein Misfolding Cyclic Amplification)<br />

4 th round product CDI (Conformation Dependent Immunoassay)<br />

Sensitivity: 10 -8 dilution of brain homogenate and 10 -5 of spleen homogenate in<br />

plasma<br />

Evaluation:<br />

1) 250 normal UK donor samples. One PRNP-129MM (No45) repeat-reactive.<br />

Positive, negative controls and No45 products inoculated tg650 mice + and 45<br />

spleen accumulation, some clinical disease (completed April 2012).<br />

2) 400 normal US plasma samples. Spiked with brain and spleen homogenates<br />

(NIBSC), 20 weeks to complete. Request for clinical samples depending on<br />

satisfactory evaluation data.<br />

Alliance BioSecure project:<br />

1) Select the best assay (most efficient plasma pre-treatment method and PMCA<br />

substrate; most sensitive PrP Sc detection method).<br />

Pre-treatment: NaCl precipitation ~ magnetic beads capture ~ streptomycin<br />

precipitation.<br />

Detection: CDI > Western blotting.<br />

Ongoing evaluation and validation.<br />

2) PrP Sc amplification = amplification of infectivity?<br />

Products of spiked samples inoculated into tg650 mice ~ 2 years to complete.<br />

B) Lyme borreliosis<br />

Lyme borreliosis (Lyme Disease) is caused by the spirochete Borrelia burgdorferi<br />

transmitted in Scotland by the Ixodes ricinus tick. Increased incidence was described in<br />

recent years, with Highlands being a hot spot, although the prevalence in the <strong>Scottish</strong><br />

population is generally unknown.<br />

NMRU and <strong>National</strong> Lyme Borreliosis testing Laboratory (Inverness): 706 donor samples<br />

tested, 5.7 %. Study currently being extended to samples from east of Scotland (~700), as<br />

an increased incidence was indicated in Tayside.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 29 of 74


(Estimated amount of impact on patient/population: small +;medium ++ ;high +++)<br />

Project Safe Effecti- Efficient Equitable Comment<br />

ve<br />

1a +++ +++ ++ ++ It is not known if WNV, Dengue, Chikungunya, Usutu viruses,<br />

Trypanosoma parasite can get established in the UK, or on a larger<br />

scale in Europe tourist destinations. Preparedness for such eventuality<br />

in a form of having risk assessments and evaluated working assays is<br />

necessary, since the impact on the UK blood supply could be<br />

significant.<br />

1b-I +++ ++ ++ ++ A proportion of human papillomaviruses, polyomaviruses, herpes v.<br />

6,7,8 or parvovirus B19 which originate in blood or blood products and<br />

may harm immunosuppressed recipients is judged minor in<br />

comparison with activation (except B19), but not known. Preventative<br />

measures, if practicable should improve the post-transplant outcomes<br />

and decrease/eliminate blood services’ liability.<br />

1b-II +++ ++ + ++ Determining the <strong>Scottish</strong> prevalence figures for HEV should indicate if<br />

any form of (partial) screening may be necessary. HEV is known to<br />

cause a chronic infection in immunosuppressed.<br />

1c ++ + + ++ Mutants represent a danger for any screening assay. HBsAg is the<br />

most common assay for HBV and some interference of the assay<br />

caused by mutants and vaccination is known. Improvements by<br />

inclusion of additional monoclonal antibodies could be beneficial.<br />

2a +++ ++ ++ ++ Non-enveloped viruses pose problem for pathogen reduction<br />

technologies (PRT). It may be easier to remove, rather than inactivate<br />

them. Synthetic polymers are good candidate materials for this<br />

purpose. Partnering with commercial partner is envisaged.<br />

2b +++ +++ + ++ Combination of new diagnostic technologies with PRT seems most<br />

feasible for future improvements of blood safety. Niche market<br />

applications such as blood screening have to be addressed largely by<br />

blood establishments as they are usually too small for exclusive<br />

commercial development. Microarrays, new nucleic acid testing<br />

methods and next generation sequencing are intertwined in much<br />

improved future diagnostics. Joint development with commercial<br />

partner can bring IP, income generation.<br />

3a-I-III ++ +++ +++ ++ Materials (PAS5) and techniques (inhibition of activation, targeting new<br />

molecules involved in activation) for reduction of platelet storage lesion<br />

and extension of shelf life are an important part of improving the blood<br />

component quality, benefiting the patients.<br />

3b ++ ++ + + All important question of the effects of fresh and aged blood has not<br />

been resolved yet and new approaches facilitating clarification are<br />

important. Use of synthetic polymers for separation and/or assaying<br />

fresh and aged subpopulations complements other R&D projects in<br />

addressing this problem.<br />

4a-c +++ ++ +++ ++ Successful validations are important precondition of translation of R&D<br />

into practice and require input from both, R&D and Supply chain. O/n<br />

hold may impact substantailly on delivery of blood and blood<br />

components from manufacturing (P&T) sites to clinical banks,<br />

improving the logistics of the process. PRT is expected to contribute<br />

substantially to blood safety. Better additive solutions would impact on<br />

the quality and shelf life of blood components.<br />

5a ++ +++ +++ + Good confirmatory vCJD assay is essential for a potential introduction<br />

of the screening assay, to avoid excessive blood wastage.<br />

5b ++ ++ + ++ Prevalence estimation for Borrelia burgdorferi will serve as a guide for<br />

potential future measures.<br />

Project Milestones<br />

(TBC = To be completed by)<br />

Project<br />

1<br />

Epidemiology<br />

pathogenesis<br />

& diagnostics<br />

of blood<br />

borne<br />

pathogens<br />

Objective<br />

Evaluate available<br />

commercial or inhouse<br />

assays for<br />

emerging blood<br />

borne pathogens in<br />

response to SACTTI<br />

Yr 1<br />

04/12-<br />

03/13<br />

Compare<br />

candidate<br />

confirmatory<br />

PCR assays<br />

for West<br />

Nile virus<br />

Yr 2<br />

04/13-<br />

03/14<br />

Complete<br />

evaluation<br />

of<br />

confirmatory<br />

PCR assay<br />

for West<br />

Milestones<br />

Yr 3<br />

04/14-<br />

03/15<br />

Yr 4<br />

04/15-03/16<br />

Yr 5<br />

04/16-03/17<br />

Develop/evaluate assays in response to SACTTI<br />

prioritisation (Dengue, Chikungunya, Usutu,<br />

Chagas)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 30 of 74


priorities<br />

Mar 2017<br />

Evaluation of the<br />

proportion of<br />

patients acquiring<br />

common viruses<br />

harmful to<br />

immunocompromis<br />

ed recipients<br />

originating from<br />

blood products<br />

Mar 2017<br />

Determination of<br />

hepatitis E virus<br />

seroprevalence and<br />

RNA frequency in<br />

<strong>Scottish</strong> blood<br />

donors and its<br />

threat to blood<br />

supply<br />

TBC Dec<br />

12<br />

Submit new<br />

applications<br />

for funding<br />

TBC<br />

June12;<br />

Oct 12<br />

Complete<br />

experimenta<br />

l part of the<br />

project to<br />

determine<br />

HEVantibody<br />

&<br />

RNA<br />

prevalence<br />

in <strong>Scottish</strong><br />

blood<br />

donors<br />

Nile virus<br />

TBC May<br />

13 Rolling - TBC Mar 2017<br />

Carry out systematic Determine the proportion<br />

collaborative study of transfusion-transmitted (rather than<br />

effects on transplant reactivated) viruses for relevant<br />

recipients. Determine combinations of virus – transplant<br />

which viruses are relevant recipient<br />

for particular transplant<br />

recipient groups.<br />

Determine prevalences in<br />

blood donors.<br />

TBC Mar 2017<br />

TBC Mar 2015<br />

Produce<br />

final report<br />

and submit<br />

publication.<br />

Submit<br />

funding<br />

application<br />

Carry out chronic hepatitis<br />

study in parallel with<br />

immunosuppressed study<br />

(above)<br />

2<br />

New<br />

diagnostic &<br />

pathogenreduction<br />

technologies<br />

March 2015<br />

Identification of<br />

synthetic polymer<br />

ligands for difficultto-inactivate<br />

viruses, for their<br />

potential removal<br />

TBC Nov<br />

12<br />

Produce<br />

final report<br />

and submit<br />

an<br />

application<br />

for<br />

extended<br />

funding<br />

TBC Jun 13<br />

Identify<br />

potential<br />

commercial<br />

partners<br />

Rolling Mar 2015<br />

Participation in development of<br />

virus removal filtration/capture<br />

material/device<br />

Feb 2016<br />

Evaluation of the<br />

suitability of new<br />

diagnostic<br />

platforms for blood<br />

testing and safetymargin<br />

increase<br />

TBC Jan 13<br />

Prepare and<br />

submit<br />

business<br />

case for<br />

funding to<br />

evaluate<br />

next<br />

generation<br />

sequencing<br />

for safety<br />

and QC<br />

testing<br />

TBC Jul 13<br />

Pilot study<br />

on<br />

diagnostics<br />

of bloodborne<br />

pathogens.<br />

Rolling<br />

Pilot study on use of the<br />

technology to QC stem cell<br />

derived lines/ products<br />

(with JM – Tissue & Cellular<br />

Therapies)<br />

Comparison with<br />

approved testing<br />

methods and<br />

cost-efficiency<br />

evaluation<br />

3<br />

Improvement<br />

s in quality &<br />

safety of<br />

blood<br />

components<br />

To identify the best<br />

available platelet<br />

and red cell additive<br />

solutions and<br />

supplements for<br />

optimal quality of<br />

blood components<br />

Mar 2017<br />

TBC Dec<br />

12<br />

Publish<br />

results of<br />

PAS 5 study<br />

TBC Nov<br />

12<br />

TBC Mar 14<br />

Evaluate<br />

feasibility of<br />

PAS 5<br />

validation in<br />

conjunction<br />

with<br />

pathogen<br />

reduction<br />

Rolling<br />

Carry out<br />

SOLX<br />

evaluation<br />

Rolling<br />

TBD – further studies on additive<br />

solutions<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 31 of 74


Identification of an<br />

improved storage<br />

protocol to prevent<br />

activation of<br />

platelets<br />

Identify best<br />

solvent for<br />

the inhibitor,<br />

not<br />

interfering<br />

with platelet<br />

activation<br />

TBC Mar 14 TBC Mar 15 Rolling<br />

Investigatio N/A<br />

n of<br />

beneficiary<br />

effect of a<br />

MAPK<br />

kinase –<br />

inhibitor on<br />

platelet<br />

storage<br />

lesion<br />

Dec 2014<br />

Determination of<br />

key platelet<br />

activation pathways<br />

using natural and<br />

synthetic activators<br />

TBC Mar 13<br />

Submit<br />

paper<br />

analysing<br />

miRNA<br />

profiles.<br />

Submit<br />

grant<br />

application.<br />

TBC Dec<br />

14<br />

Correlate<br />

miRNA data<br />

with gene<br />

expression<br />

studies<br />

Select the<br />

key<br />

activation<br />

molecules<br />

Investigate<br />

potential<br />

application to<br />

storage lesion<br />

4<br />

Validation<br />

studies for<br />

Components<br />

&<br />

Microbiology<br />

Feb 2016<br />

Validation of<br />

overnight hold &<br />

pathogen reduction<br />

technologies in<br />

collaboration with<br />

Supply Chain<br />

TBC Mar 13<br />

TBC Mar 14 TBC Mar 15<br />

Perform required tests for<br />

validation<br />

TBC Feb 16<br />

5<br />

Non-Viral<br />

Microbiology<br />

research<br />

March 2014<br />

Validation of vCJD<br />

confirmatory<br />

assay<br />

Mar 2014<br />

Development of an<br />

assay to detect<br />

active Borrelia<br />

burgdorferi<br />

infection<br />

Complete<br />

optimisation<br />

of PMCA<br />

and initial<br />

evaluation<br />

with NIBSC<br />

blinded<br />

spiked<br />

plasma<br />

panel<br />

TBC Mar 13<br />

Design<br />

primers and<br />

probes and<br />

optimise<br />

PCR assay<br />

March 2014<br />

Complete<br />

validation<br />

of PMCA<br />

test<br />

for vCJD<br />

TBC Mar 14<br />

Carry out<br />

pilot study<br />

Report on<br />

assay<br />

suitability;<br />

SACTTI risk<br />

assessment<br />

and SABTO<br />

position<br />

statement<br />

June 2014<br />

Mar 13<br />

TBC Mar 14<br />

TBC June<br />

14<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 32 of 74


Total resources allocated FY 2012-13<br />

Project Staff Cost G&S SRC TOTAL<br />

MC1 Epidemiology,<br />

£138,563 £23,650 £5,710 £167,923<br />

pathogenesis & diagnostics<br />

of blood borne pathogens<br />

MC2 New diagnostic &<br />

£147,544 £39,205 £5,910 £192,659<br />

pathogen-reduction<br />

technologies<br />

MC3 Improvements in<br />

£106,936 £36,310 £7,640 £150,886<br />

quality & safety of blood<br />

components<br />

MC4 Validation studies for £69,694 £8,578 £4,149 £73,886<br />

Components &<br />

Microbiology<br />

MC5 Non-Viral<br />

Microbiology research<br />

NMRU<br />

(NMRU)<br />

TOTAL £462,737 £107,743 £23,409 £593,889<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 33 of 74


Clinical <strong>Transfusion</strong> Appendix 4<br />

Over the next five years, the theme will concentrate most of its resource on evaluating and<br />

minimising adverse effects of blood transfusion in light of the accumulating evidence that<br />

excessive transfusion especially of aged cells causes excess morbidity and mortality in<br />

patients. This may be a ‘silent epidemic’ in which case we ought to be investigating it and<br />

carrying out mitigating actions where possible. There are also a number of diagnostic and<br />

therapeutic development projects which we wish to transition to the commercial sector.<br />

The underlying strategy will be to link pre-existing expertise into specific subject areas<br />

where it is judged we can become internationally competitive.<br />

Expected Achievements [Outputs]:<br />

1. To determine whether red cell transfusion can increase mortality in sick patients.<br />

2. To understand whether and how uptake of transfused blood by macrophages may<br />

induce inappropriate activation of the immune system.<br />

3. To determine whether and how red cell transfusion may induce inappropriate<br />

activation of coagulation.<br />

4. To investigate determinants of red cell alloantibody formation.<br />

5. To provide support for molecular diagnostic services.<br />

6. To commercialise array-based diagnostics (‘mimotopes’).<br />

7. To commercialise peptide based therapies, directed initially against RhD.<br />

8. To test immunological properties of erythrocytes produced from <strong>Blood</strong>pharma.<br />

9. To monitor HLA sensitisation after cell therapies provided by <strong>SNBTS</strong>.<br />

10. To develop human monoclonal antibodies against red cell antigens.<br />

11. To test new therapeutic antibodies using placental transport models.<br />

12. To measure haematopoietic stem cell kinetics after transplanation.<br />

Benefits to <strong>SNBTS</strong> and NHS Scotland [Outcomes]:<br />

1. To optimise clinical decisions concerning the appropriateness of blood transfusion.<br />

2. To optimise the duration and conditions of blood storage.<br />

3. To improve the safety of cell products provided by <strong>SNBTS</strong>.<br />

4. To generate income from new diagnostic and therapeutic materials.<br />

1. To determine whether red cell transfusion increases mortality in sick patients.<br />

This project will comprise support for three clinical trials:<br />

A. The REstrictive and LIberal transfusion strategies in intEnsiVE care (RELIEVE) study.<br />

B. The Age of <strong>Blood</strong> Evaluation (ABLE) study.<br />

C. The <strong>Transfusion</strong> in Gastrointestinal Bleeding (TRIGGER study).<br />

The background to these trials is that blood transfusion has been previously assumed to be<br />

clinically beneficial to anaemic patients in most clinical situations, but has not been<br />

subjected to rigorous, modern testing in the same way that drugs or other clinical<br />

interventions have been. Some studies have suggested that liberal transfusion regimens<br />

are associated with higher morbidity and mortality rates then conservative ones in some<br />

patient groups and that aged red cell concentrates may be associated with higher rate of<br />

adverse outcomes than younger red cell concentrates. It is important to establish therefore<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 34 of 74


oth whether these initial findings are correct and the potential mechanisms thereof in order<br />

to guide further policy.<br />

More specifically:<br />

A. The RELIEVE study will test whether transfusion triggers of 70 vs 90g/L are associated<br />

with better / worse outcomes in patients requiring ventilation in intensive care. Current<br />

pilot data indicate that a more liberal transfusion strategy is associated with a higher<br />

mortality. It is therefore critical to determine whether this effect is true.<br />

B. The ABLE study addresses whether ‘older’ blood increases mortality. <strong>Blood</strong> is currently<br />

stored for up to 35 days before it is considered out of date, a figure determined mainly<br />

from in vitro studies. Several recent studies have suggested that recipients of blood that<br />

has been stored for longer periods have a higher mortality than those receiving younger<br />

blood. It is important to note that these studies had design faults and other, apparently<br />

similar, studies have not confirmed the effects. If the effects are confirmed, this would<br />

suggest that red cell transfusion, as currently practiced, may be contributing to excess<br />

morbidity / mortality in some patient groups. It is therefore important that this effect is<br />

investigated in a large, prospective, randomised trial. We feel it is important that<br />

Scotland takes part in the international ABLE study.<br />

C. The TRIGGER study will address what threshold should be used for red cell<br />

transfusions in acute upper gastrointestinal bleeding (AUGIB), one of the leading<br />

indications for transfusions. This is a UK based multi-centre cluster randomised<br />

controlled trial with Edinburgh as one of the six centres.<br />

Resources to be allocated:<br />

It is proposed that co-ordination of these three trials will be performed by <strong>SNBTS</strong> Product<br />

Services Department.<br />

For these three trials, the first milestone will be recruitment of first <strong>Scottish</strong> patients and this<br />

is scheduled for April 2013.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 35 of 74


2. To understand whether and how uptake of transfused blood by macrophages<br />

may induce inappropriate activation of the immune system.<br />

The recent evidence implicating older stored red cells as contributors to mortality highlights<br />

the surprising lack of knowledge we have about how red cells are marked for destruction in<br />

vivo and the immunological implications of the different modes of death. It has long been<br />

known that up to 25% of red cells from older bags of blood are removed from circulation<br />

within one hour of being transfused, by splenic and hepatic macrophages. Data from<br />

animals indicate that those cells cause inappropriate activation of the immune system and<br />

subsequent death. It is therefore proposed to determine which of several proposed<br />

mechanisms mark red cells that have come to the end of their lives and the responses of<br />

macrophages that take up these effete red cells.<br />

This work will proceed with the support of a Wellcome Trust funded programme grant. The<br />

<strong>SNBTS</strong> contribution will comprise several strands:<br />

(A) Optimise an in vitro macrophage uptake assay.<br />

(B) Assess the responses of macrophages that have taken up red cells using a<br />

transcriptomic approach.<br />

(C) Link with the <strong>Blood</strong> Pharma erythroid differentiation system to knock down specific<br />

red cell proteins, notably CD47, to test their functional effects on red cell uptake. We<br />

will also test whether the erythrocytes resulting from the <strong>Blood</strong> Pharma project are<br />

taken up by macrophages in a similar fashion to normally derived red cells.<br />

Resources to be allocated:<br />

Strand A: One non-tenured post-doctoral scientist (LC) for 5 years.<br />

Strand B: One tenured post-doctoral scientist (MM) for 5 years.<br />

Strand C: 30% of Glasgow team for 2 years (DC, AM, DD)<br />

3. To determine whether and how red cell transfusion may induce inappropriate<br />

activation of coagulation.<br />

A further way in which red cells may cause harm is to stimulate inappropriate coagulation.<br />

Our approach here will be firstly, to titrate red cells into a coagulation system that allows for<br />

their introduction (thromboelastography (TEG)) and measure thrombin production.<br />

Secondly, we will assay the presence of prothrombin surface markers using flow cytometry.<br />

Resources to be allocated:<br />

£5,000 for consumables in Dundee (TEG)<br />

£5,000 for consumables in Edinburgh (FACS)<br />

0.5wte for salary in Edinburgh (SM/ID)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 36 of 74


4. To determine whether HLA type determines propensity to red cell alloantibody<br />

formation.<br />

The formation of alloantibodies against blood remains a significant clinical problem, which<br />

complicates and may delay the provision of appropriately matched cells. A study will be<br />

undertaken to examine the relationship between HLA alleles and RBC alloantibody<br />

formation in the West of Scotland patient population.<br />

Resources to be allocated: 5% of AM’s time for 2 years, to collect samples. Consumables<br />

funding will depend on successful grant application and if this is not obtained, project will be<br />

abandoned.<br />

5. To provide support for molecular diagnostic services.<br />

Specialised red cell, platelet and granulocyte genotyping services, including prenatal<br />

diagnosis, benefit from the input of senior R&D staff. The support encompasses both<br />

troubleshooting and the development of new diagnostic tests (prenatal diagnosis of anti-<br />

HPA mediated diseases, diagnosis of PNH).<br />

Resources to be allocated: 0.2 FTE for 5 years (SAF)<br />

6. To commercialise array-based diagnostics (‘mimotopes’).<br />

The project has been taken beyond “proof of concept” to proof of potential utility and is well<br />

positioned to be taken on by a commercial entity to further develop (in combination with an<br />

existing platform) to a marketable product / system.<br />

For this reason all <strong>SNBTS</strong> developments are on hold. However, the Principal Investigator<br />

Mike Moss remains available to support, to a limited degree, a third party development<br />

endeavour and this input will be critical in progressing this project into a commercial arena.<br />

Resources to be allocated: 0.1 FTE for 2 years (MM).<br />

7. To commercialise peptide based therapies, directed initially against RhD.<br />

No further laboratory work is currently planned on this project. However, input of scientific<br />

staff will be critical to progress this project into a commercial arena.<br />

Resources to be allocated: 0.1 FTE for 2 years (LC)<br />

8. To test immunological properties of erythrocytes produced from <strong>Blood</strong>pharma.<br />

The red cells to be produced from the <strong>Blood</strong> Pharma project will need to be rigorously<br />

tested for equivalence to their normal counterparts, before studies in humans. We will<br />

utilise the assays developed as part of project 2 to test the surface properties and<br />

immunological effects of these red cells.<br />

Resources to be allocated: 0 in years 1 & 2, contribution in year 3 dependent on<br />

<strong>Blood</strong>Pharma project progress.<br />

9. To monitor HLA sensitisation after cell therapies provided by <strong>SNBTS</strong>.<br />

This project will support two treatments arising from the Cellular Therapy programme (islet<br />

cell transplantation and Epstein-Barr virus cytotoxic lymphocytes (EBV-CTLs)) by<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 37 of 74


establishing whether the presence of anti-HLA antibodies in recipients correlates with<br />

clinical outcome. Also, it is proposed to identify donors homozygote for common HLA<br />

haplotypes in a <strong>Scottish</strong> population, for subsequent development of iPS cells. Cross refer to<br />

relevant CT projects.<br />

Resources to be allocated:<br />

Year 1: £10,500 for HLA typing<br />

Years 2-4: £11,200 per annum for anti-HLA antibody detection.<br />

DN: you’ve identified the money for this within budget?<br />

10. To develop human monoclonal antibodies against red cell antigens.<br />

A monoclonal antibody against HbH would be useful for prenatal diagnosis of<br />

thalassaemias in ethnic minority populations.<br />

Resources to be allocated:<br />

Year 1: £5,000 consumables + 0.2FTE of AM time in Glasgow.<br />

KPI: If no suitable antibody has been isolated in the first year, the project will be abandoned.<br />

11. To test new therapeutic antibodies using placental transport models.<br />

<strong>SNBTS</strong> has been involved in testing the properties of three antibodies that have the<br />

potential to be useful in the treatment of haemolytic disease of the newborn and neonatal<br />

alloimmune thrombocytopenia.<br />

Resources to be allocated:<br />

No specific resource is planned to support these antibodies, 0.1FTE of SAF’s time for 2<br />

years.<br />

12. To measure haematopoietic stem cell kinetics after transplanation<br />

By measuring the presence of mutant red cells, the behaviour of stem cells collected and<br />

transplanated by <strong>SNBTS</strong> can be deduced.<br />

Resources to be allocated:<br />

0.1FTE of MM’s and SH’s time for 5 years.<br />

HEALTHCARE QUALITY IMPACT ASSESSMENT (Appendix 6)<br />

(Estimated amount of impact on patient/population: small +;medium ++ ;high +++)<br />

STUDY SAFE EFFECTIVE EFFICIENT EQUITABLE<br />

ABLE +++ ++ ++<br />

1 RELIEVE +++ ++ ++<br />

TRIGGER +++ ++ ++<br />

2 Red cell signalling +++ + ++<br />

Macrophage response<br />

3 Red cells and ++ +<br />

coagulation<br />

4 Red cell alloantibodies ++ +<br />

5 Molecular diagnostic + +++ +<br />

support<br />

6 Mimotopes + +++ ++<br />

7 Peptide therapies for<br />

+++ +<br />

HDN<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 38 of 74


8 <strong>Blood</strong> Pharma testing + +++<br />

9 Cellular therapies HLA ++ ++<br />

testing<br />

10 Anti-HbH development ++ ++ ++<br />

11 Placental transport of<br />

+++<br />

antibodies<br />

12 To measure<br />

haematopoietic stem cell<br />

kinetics posttransplanation<br />

+ + +<br />

None of these projects is expected to impact on timeliness person centeredness<br />

Project Milestones<br />

(TBC = To be completed by)<br />

Project<br />

1<br />

Determine<br />

whether red<br />

cell<br />

transfusion<br />

increases<br />

mortality in<br />

sick patients<br />

Objective<br />

Participate in ABLE<br />

study (multi-centre<br />

trial comparing<br />

outcomes of<br />

transfusing fresh vs<br />

standard RBC in<br />

ICU)<br />

Yr 1<br />

04/12-<br />

03/13<br />

Yr 2<br />

04/13-03/14<br />

Start patient recruitment to study<br />

Milestones<br />

Yr 3<br />

04/14-<br />

03/15<br />

Yr 4<br />

04/15-03/16<br />

Complete patient<br />

recruitment and<br />

analyse data<br />

Yr 5<br />

04/16-03/17<br />

Dec 2015<br />

Carry out RELIEVE<br />

study (comparing<br />

restrictive vs liberal<br />

transfusion in<br />

intensive care)<br />

Apr 13 and ongoing<br />

Publish Initiate<br />

internal RELIEVE<br />

feasibility study multicentre<br />

and plan larger study and<br />

scale trial complete<br />

recruitment of<br />

25 patients at<br />

6 centres by<br />

April 2013<br />

TBC Dec 15<br />

Continue patient recruitment to study<br />

Participate in<br />

TRIGGER study<br />

(comparing<br />

restrictive vs liberal<br />

transfusion in GI<br />

bleeding)<br />

TBC Dec 12<br />

Start patient<br />

recruitment<br />

TBC Apr 13<br />

Initiate scaleup<br />

studies<br />

incorporating<br />

new partners<br />

if necessary<br />

Rolling<br />

2<br />

Red cell<br />

signalling<br />

macrophage<br />

response<br />

Mar 2016<br />

Investigate the<br />

effect of transfused<br />

blood on the<br />

immune system<br />

(5yr study)<br />

Oct 12<br />

Complete<br />

recruitment of<br />

volunteers/pati<br />

ents and<br />

complete<br />

protocol<br />

design. Isolate<br />

macrophages<br />

that have<br />

taken up red<br />

cells for<br />

Rolling<br />

Continue investigation on effects of aged RBC uptake on human<br />

phagocytic cells<br />

Investigate how accumulation of ‘storage lesions’ in blood for<br />

transfusion alters the immune effects of RBC clearance<br />

Determine the role of CD47 in RBC senescence<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 39 of 74


transcriptomic<br />

analysis by<br />

Mar 2017<br />

Perform<br />

comprehensive<br />

glycomic analysis<br />

of red cells stored<br />

for transfusion<br />

Aug 12. Rolling - TBC Mar 17<br />

Establish panel<br />

of linkage<br />

specific<br />

glycosyl<br />

binding<br />

molecules.<br />

Establish mass<br />

spectroscopic<br />

analysis<br />

techniques in<br />

partnership<br />

with UCL<br />

Test stored<br />

blood for<br />

glysosylation<br />

patterns using<br />

reagents &<br />

techniques<br />

established<br />

from 2012/13<br />

3<br />

Red cells and<br />

coagulation<br />

4<br />

Red cell<br />

alloantibodies<br />

Assess<br />

immunological<br />

consequences of<br />

red cell uptake by<br />

splenic/hepatic<br />

macrophages<br />

Investigate how red<br />

cell transfusion<br />

may induce<br />

inappropriate<br />

activation of<br />

coagulation<br />

March 2014<br />

Determine if HLA<br />

type determines<br />

propensity to red<br />

cell alloantibody<br />

formation<br />

Mar 13<br />

Establish<br />

assay for take<br />

up of red cells<br />

by<br />

macrophages<br />

Mar 13<br />

Plan<br />

experiments to<br />

determine<br />

effects of red<br />

cells on<br />

clotting by<br />

thromboelasto<br />

graphy and<br />

assess<br />

expression of<br />

coagulation<br />

markers on red<br />

cells by flow<br />

cytometry<br />

TBC Mar 13<br />

Submit ethics<br />

and apply for<br />

grant funding<br />

Mar 14<br />

Perform transcriptomic analyses<br />

of macrophage responses to<br />

transfused cells<br />

Mar 15<br />

Initiate<br />

experimental<br />

work<br />

Rolling<br />

Collect patient<br />

samples and<br />

carry out study<br />

Complete<br />

experimental<br />

work and<br />

submit for<br />

publication<br />

TBC Mar 14<br />

Submit for<br />

publication<br />

Submit for<br />

publication<br />

Mar 16<br />

5<br />

Molecular<br />

diagnostic<br />

support<br />

March 2014<br />

Implement noninvasive<br />

prenatal<br />

diagnosis (NIPD)<br />

for Rh status in<br />

pregnancy<br />

(screening)<br />

TBC Mar 13<br />

Complete<br />

implementation<br />

trial for NIPD<br />

TBC Mar 14<br />

TBC Mar 15<br />

6<br />

Mimotopes<br />

7<br />

Peptide<br />

therapies<br />

HDN<br />

for<br />

Dec 2013<br />

Commercialise<br />

array-based<br />

diagnostics<br />

(‘mimitopes’)<br />

Commercialise<br />

peptide based<br />

therapies, directed<br />

TBC Mar 13<br />

Continue to negotiate with prospective commercial<br />

partners. No further experimental work planned<br />

unless funded by potentially interested commercial<br />

partner<br />

Rolling<br />

Continue to negotiate with prospective commercial<br />

partners. No further experimental work planned<br />

unless funded by potentially interested commercial<br />

partner<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 40 of 74


8<br />

<strong>Blood</strong> Pharma<br />

testing<br />

9<br />

Cellular<br />

therapies HLA<br />

testing<br />

against RhD or<br />

GPIIb/IIIa<br />

Test immunological<br />

properties of<br />

erythrocytes<br />

produced from<br />

<strong>Blood</strong>Pharma<br />

March 2017<br />

Monitor HLA<br />

sensitisation after<br />

cell therapies<br />

provided by<br />

<strong>SNBTS</strong><br />

Rolling<br />

Test red cells from <strong>Blood</strong>Pharma<br />

project in macrophage uptake assay<br />

Precise timing dependent on<br />

progress of <strong>Blood</strong>Pharma<br />

project<br />

Collect and analyse samples when required. Timing depends on progress of<br />

various cellular therapy projects.<br />

10<br />

Anti-HbH<br />

development<br />

Dec 2016<br />

Generate a murine<br />

monoclonal<br />

antibody against<br />

HbH<br />

Rolling<br />

Perform<br />

mouse<br />

immunisation,<br />

fusion,<br />

screening,<br />

cloning and<br />

characterisatio<br />

n of antibodies<br />

Project<br />

terminates<br />

11<br />

Placental<br />

transport of<br />

antibodies<br />

March 2013<br />

Obtain external<br />

funding for testing<br />

new therapeutic<br />

antibodies using<br />

placental transport<br />

models<br />

TBC Mar 13<br />

Continued experimental<br />

work dependent on specific<br />

externally-funded projects<br />

12<br />

Measure<br />

haematopoieti<br />

c stem cell<br />

kinetics posttransplanation<br />

May 2014<br />

Measure<br />

haematopoietic<br />

stem cell kinetics<br />

after transplanation<br />

Rolling<br />

Collect and freeze samples<br />

Analyse<br />

samples-may<br />

need external<br />

funding<br />

Total resources allocated FY 2012-13<br />

Theme Project No Staff Cost G&S SRC TOTAL<br />

1 Non-RDI Staff<br />

2 £184,507 £5,488 £13,744 £203,729<br />

3 £61,048 £6,567 £67,615<br />

Improve Efficacy &<br />

Safety of <strong>Blood</strong><br />

<strong>Transfusion</strong> for Patients<br />

in Scotland<br />

Development of<br />

Diagnostic and<br />

Therapeutic<br />

Approaches for<br />

Diseases of <strong>Blood</strong><br />

4 £5,235 £693 £1,566 £7,494<br />

5 £44,515 £3,190 £9,457 £57,162<br />

6 £10,168 £1,109 £2,882 £14,159<br />

7 £8,177 £694 £1,567 £10,438<br />

8<br />

9 £23,951 £1,942 £5,512 £31,405<br />

10 £9,698 £693 £1,000 £11,391<br />

11 £10,168 £1,110 £2,882 £14,160<br />

12 £10,168 £1,110 £2,882 £14,160<br />

TOTAL £367,635 £16,019 £48,059 £431,713<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 41 of 74


Publications & Grants 2008-2012 Appendix 5<br />

2008-2009<br />

Aldhous, M. C., Prescott, R. J., Roberts, S., Samuel, K., Waterfall, M., and Satsangi, J. Does nicotine<br />

influence cytokine profile and subsequent cell cycling/apoptotic responses in inflammatory bowel disease?<br />

Inflamm Bowel Dis. 2008: 14: 1469-1482.<br />

Appleford NE, Wilson K, Houston F, Bruce LJ, Morrison A, Bishop M, Chalmers K, Miele G, Massey E, Prowse<br />

C, Manson J, Will RG, Clinton M, MacGregor I, Anstee DJ. alpha-Hemoglobin stabilizing protein is not a<br />

suitable marker for a screening test for variant Creutzfeldt-Jakob disease. <strong>Transfusion</strong>. 2008; 48: 1616-26.<br />

Balabanov S, Gontarewicz A, Ziegler P, Hartmann U, Kammer W, Copland M, Brassat U, Priemer M, Hauber<br />

I, Wilhelm T, Schwarz G, Kanz L, Bokemeyer C, Hauber J, Holyoake TL, Nordheim A, Brummendorf TH.<br />

Hypusination of eukaryotic initiation factor 5A (EIF05A): a novel therapeutic target in BCR-ABL-positive<br />

leukemias identified by a proteomics approach. <strong>Blood</strong> 2007, 109: 1701-1711.<br />

Bateman AP, McArdle F, Walsh TS. Time course of anemia during six months follow up following intensive<br />

care discharge and factors associated with impaired recovery of erythropoiesis. Crit Care Med. 2009<br />

Jun;37(6):1906-12<br />

Beattie LM, Harrison E “SAGM blood for neonatal large volume transfusion.” <strong>Transfusion</strong> Alternatives in<br />

<strong>Transfusion</strong> Medicine 2008; 10: pp 34-36<br />

Bessos H, Matviyenko M, Brown P, Husebekk A, Killie MP, Segatchian J, Santoso S, Urbaniak SJ What’s<br />

happening – probing of HPA-1a antigen-antibody interaction by surface plasmon resonance technology.<br />

<strong>Transfusion</strong> and Apheresis Science (2008), 39; 179-182<br />

Bessos H, Matviyenko M, Killie MK, Husebekk A, Urbaniak S. Direct comparison between ELISA and MAIPA<br />

in the measurement of anti-HPA-1a (in IU/ml and AU/ml) in Neonatal Alloimmune Thrombocytopenia.<br />

<strong>Transfusion</strong> and Apheresis Science (2008), 39, 221-227<br />

Bessos H, Killie MK, Seghatchian J, Skogen B, Urbaniak SJ. What's Happening - The Relationship of Anti-<br />

HPA-1a Amount to Severity of Neonatal Alloimmune Thrombocytopenia - Where Does it Stand? Transfus<br />

Apher Sci. 2009; 40: 75-8<br />

Brazma D, Grace C, Howard J, Melo JV, Holyoake T, Apperley JF, Nacheva EP. Genomic Profile of CML:<br />

Imbalances Associated with Disease Progression. Genes, Chromosomes and Cancer, 2007: 46(11) 1039-59.<br />

Brunskill SJ, Prowse C, Garrioch M, Gill R, Hebert P, Thompson J, Hyde C, Stanworth S, Roberts D. <strong>Blood</strong><br />

substitutes for avoiding allogeneic blood transfusion (Protocol)The Cochrane Library 2008, Issue 1, pp1-6.<br />

Burgess STG, Kenyon F, O’Looney N, Ross AJ, Chong Kwan M, Beattie J, Petrik J, Ghazal P, Campbell C: A<br />

multiplexed protein microarray for the simultaneous serodiagnosis of HIV/HCV infection and typing of whole<br />

blood. Analytical Chemistry 2008; 382:9-15.<br />

Cardigan R, Prowse CV, Williamson LM. Processing of Components: leucodpeletion and pathogen reduction.<br />

Chapter 19 in “<strong>Transfusion</strong> Microbiology”, editors Barbara JAJ, Regan FAM, Contreras MC; publishers<br />

Cambridge University Press. 2008. pp239-258.<br />

Copland M, Hamilton A, Holyoake TL. Conventional Western blotting techniques will not reliably quantify p210<br />

BCR-ABL. <strong>Blood</strong>, (letter) 2007: 109 (3): 1336.<br />

Copland M, Pellicano F, Richmond L, Allan EK, Hamilton A, Lee FY, Weinmann R, and Holyoake TL. (2008)<br />

BMS-214662 potently induces apoptosis of chronic myeloid leukemia stem and progenitor cells and<br />

synergizes with tyrosine kinase inhibitors. <strong>Blood</strong>, 2008; 111(5):2843-53.<br />

Coste J, Prowse C, Eglin D, Fang C. A Report on Transmissible Spongiform Encephalopathies and<br />

<strong>Transfusion</strong> Safety. Vox Sanguinis 2009; 96: 284-291.<br />

Davidson F, Yirrell D, Lycett C, Petrik J, Dow BC. HIV subtypes detected in <strong>Scottish</strong> blood donors. Vox<br />

Sanguinis 2009 ;96:160-2<br />

Duncan C, Roddie H. Dendritic cell vaccines in Acute Leukemia. Best Practice & <strong>Research</strong> Clinical<br />

Haematology 2008 21(3) 521-41<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 42 of 74


Ferguson E, Prowse C, Townsend E, Spence A, Hilten JA, Lowe K. Acceptability of blood and blood<br />

substitutes. J Intern Med. 2008; 263: 244-55.<br />

Fisher SA, Tremelling M, Anderson CA, Gwilliam R, Bumpstead S, Prescott NJ, Nimmo ER, Massey D,<br />

Berzuini C, Johnson C, Barrett JC, Cummings FR, Drummond H, Lees CW, Onnie CM, Hanson CE, Blaszczyk<br />

K, Inouye M, Ewels P, Ravindrarajah R, Keniry A, Hunt S, Carter M, Watkins N, Ouwehand W, Lewis CM,<br />

Cardon L; Wellcome Trust Case Control Consortium, Lobo A, Forbes A, Sanderson J, Jewell DP, Mansfield<br />

JC, Deloukas P, Mathew CG, Parkes M, Satsangi J. Genetic determinants of ulcerative colitis include the<br />

ECM1 locus and five loci implicated in Crohn's disease. Nat Genet. 2008 ; 40: 710-2.<br />

Fletcher, J., Cui, W., Samuel, K., Black, J. R., Hannoun, Z., Currie, I. S., Terrace, J. D., Payne, C., Filippi, C.,<br />

Newsome, P., Forbes, S. J., Ross, J. A., Iredale, J. P., and Hay, D. C. The inhibitory role of stromal cell<br />

mesenchyme on human embryonic stem cell hepatocyte differentiation is overcome by Wnt3a treatment.<br />

Cloning Stem Cells 2008: 10: 331-339.<br />

Graham SM, Vass JK, Holyoake TL, Graham GJ. Transcriptional analysis of quiescent and proliferating<br />

CD34+ human haemopoietic cells from normal and CML sources. Stem Cells, 2007: 25:3111-20.<br />

Gray A, Hart M, Dalrymple K, Davies T. Promoting safe transfusion practice: right blood, right patient, right<br />

time. Br J Nurs. 2008 Jul 10-23;17(13):812, 814-7.<br />

Griffiths SD, Burthem J, Unwin RD, Holyoake TL, Melo JV, Lucas GS, Whetton AD. The use of isobaric tage<br />

peptide labelling (iTRAQ) and mass spectrometry to examine rare, primitive hematopoietic cells from patients<br />

with chronic myeloid leukemia. Molecular Biotechnology, 2007: 36(2): 81-9.<br />

Harkness M, Clark V. The use of cell salvage during obstetric procedures: an audit of Scotland's maternity<br />

units.Scott Med J. 2008;53:24-7<br />

Harkness M, Freer Y, Prescott RJ, Warner P. Implementation of NICE recommendation for a policy of routine<br />

antenatal anti-D prophylaxis: a survey of UK maternity units. Transfus Med. 2008;18:292-5<br />

Harkness M, Palmer JB, Watson D, Walsh TS. A questionnaire-based survey of perioperative blood<br />

conservation practice for revision hip arthroplasty in Scotland. Transfus Med. 2008 Oct;18(5):296-301<br />

Haque T, Wilkie GM, Jones MM, Higgins CD, Urquhart G, Wingate P, Burns D, McAulay K, Turner M, Bellamy<br />

C, Amlot PL, Kelly D, MacGilchrist A, Ghandi MK, Swerdlow AJ, Crawford DH. Allogeneic cytotoxic T cell<br />

therapy for EBV-positive post transplant lymphoproliferative disease: results of phase II multicentre clinical<br />

trial. <strong>Blood</strong> 2007; 110: 1123-31.<br />

Halsey C, Fisher C, Strathdee G, Gibson B, Holyoake T, Vyas P, Graham G. GATA1 mutational analysis in<br />

chronic myeloid leukaemia. British Journal of Haematology, 2007: 137: 375-6.<br />

Hay DC, Fletcher J, Payne C, Terrace JD, Gallagher RCJ, Snoeys J, Black JR, Wojtacha D, Hannoun Z,<br />

Pryde A, Filippi C, Currie IS, Forbes SJ, Ross JA, Newsome PN, Iredale JP. Highly efficient differentiation of<br />

hESCs to functional hepatic endoderm requires ActivinA and Wnt3a signalling. Proceedings of the <strong>National</strong><br />

Academy of Sciences of the USA 2008 105, 12301-12306.<br />

Heaney NB, Holyoake TL. Therapetuic targets in chronic myeloid leukaemia. Hematological Oncology, 2007:<br />

25: 66-75.<br />

Heaney NB, Copland M, Stewart K, Godden J, Parker AN, McQuaker IG, Smith GM, Crawley C, Shepherd P,<br />

and Holyoake TL. (2008) Complete molecular responses are achieved after reduced intensity stem cell<br />

transplantation and donor lymphocyte infusion in chronic myeloid leukemia. <strong>Blood</strong>, 2008; 111(10) 5252-5.<br />

Holyoake TL, Heaney NB. Novel treatment strategies for chronic myeloid leukaemia. Clinical Review. Hospital<br />

Pharmacy Europe. 2007: 19-20.<br />

Hornsey VS, McColl K, Drummond O, MacGregor IR, Prowse CV. Platelet storage in Fresenius/NPBI<br />

polyolefin and BTHC-PVC bags: a direct comparison. Transfus Med. 2008; 18: 223-227.<br />

Hornsey VS, McMillan L, Morrison A, Drummond O, MacGregor IR, Prowse CV. Freezing of buffy coatderived,<br />

leukoreduced platelet concentrates in 6 percent dimethyl sulfoxide. <strong>Transfusion</strong>. 2008; 48: 2508-2514<br />

Hornsey VS, Drummond O, McMillan L, Morrison A, Morrison L, MacGregor IR, Prowse CV. Cold storage of<br />

pooled, buffy-coat-derived, leucoreduced platelets in plasma. Vox Sang. 2008; 95: 26-32<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 43 of 74


Jarvis L, Becker J, Tender A, Cleland A, Queiros L, Aquiar A, Azevedo J, Aprili G, Bressan F, Torres P, Nieto<br />

S, Ursitti A, Montoro J, Vila E, Ramada C, Saldanha J. Evaluation of the Roche cobas s 201 system and<br />

cobas TaqScreen multiplex test for blood screening: a European multicenter study. <strong>Transfusion</strong>, 2008;<br />

48(9):1853-61.<br />

Jones M, Peden AH, Prowse CV, Groner A, Manson JC, Turner ML, Ironside JW, MacGregor IR, Head MW.<br />

In vitro amplification and detection of variant Creutzfeldt-Jakob disease PrP Sc . Journal of Pathology 2007; 213:<br />

21-26.<br />

Jones M, Peden AH, Yull H, Bishop MT, Prowse CV, Turner ML, Ironside JW, Head MW, MacGregor IR.<br />

Human platelets as a substrate source for the in vitro amplification of the abnormal prion protein (PrP Sc )<br />

associated with variant Creutzfeldt-Jakob disease. <strong>Transfusion</strong>. 2009: 49: 376-384.<br />

Jones M, Wight D, McLoughlin V, Norrby K, Ironside JW, Connolly JG, Farquhar CF, MacGregor IR, Head<br />

MW. An Antibody to the Aggregated Synthetic Prion Protein Peptide (PrP106-126) Selectively Recognizes<br />

Disease-Associated Prion Protein (PrP(Sc)) from Human Brain Specimens. Brain Pathol. 2009; 19: 293-302<br />

Jones M, Peden A, Wight D, Prowse C, MacGregor I, Manson J, Turner M, Ironside JW, Head MW. Effects of<br />

sporadic Creutzfeldt-Jakob disease prion protein type and PRNP codon 129 genotype in an in vitro prion<br />

protein conversion assay. NeuroReport 2008; 19: 1783-1786.<br />

Jones M, Peden AH, Wight D, Prowse C, MacGregor I, Manson J, Turner M, Ironside JW, Head MW. Effects<br />

of human PrPSc type and PRNP genotype in an in-vitro conversion assay. Neuroreport. 2008;19:1783-1786<br />

Kilpatrick, D.C. Lectin-glycoconjugate interactions in health and disease. Biochemical Society Transactions<br />

2008; 36: 1453-1456<br />

Kilpatrick DC. Stem Cell Transplantation and MBL Replacement Therapy. Current Stem Cell <strong>Research</strong> and<br />

Therapy 2008; 3: 85-87<br />

Kilpatrick DC. Mannan-binding lectin and stem cell transplantation. In: Hematopoietic Stem Cell<br />

Transplantation Advances (Karl B. Neumann, ed), 2008, pp 1-6.<br />

Konig H, Holtz M, Modi H, Manley P, Holyoake TL, Forman SJ and Bhatia R. Enhanced BCR-ABL kinase<br />

inhibition does not result in increased inhibition of downstream signaling pathways or increased growth<br />

suppression in CML progenitors. Leukaemia 2008; 22(4): 748-55.<br />

Konig H, Holyoake TL, Bhatia R. Effective and selective inhibition of chronic myeloid leukemia primitive<br />

hematopoietic progenitors by the dual Src-Abl kinase inhibitor SKI-606. <strong>Blood</strong>, 2008; 111(4): 2329-38.<br />

Larson RA, Druker BJ, Guilhot F, O'Brien SG, Riviere GJ, Krahnke T, Gathmann I, Wang Y, International<br />

Randomized Interferon vs STI571 Study Group. Imatinib pharmacokinetics and its correlation with response<br />

and safety in chronic-phase chronic myeloid leukemia: a subanalysis of the IRIS study. <strong>Blood</strong>, 2008; 111(8)<br />

4022-8.<br />

Loos RJ, Lindgren CM, Li S, Wheeler E, Zhao JH, Prokopenko I, Inouye M, Freathy RM, Attwood AP,<br />

Beckmann JS, Berndt SI; Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial, Jacobs KB,<br />

Chanock SJ, Hayes RB, Bergmann S, Bennett AJ, Bingham SA, Bochud M, Brown M, Cauchi S, Connell JM,<br />

Cooper C, Smith GD, Day I, Dina C, De S, Dermitzakis ET, Doney AS, Elliott KS, Elliott P, Evans DM, Sadaf<br />

Farooqi I, Froguel P, Ghori J, Groves CJ, Gwilliam R, Hadley D, Hall AS, Hattersley AT, Hebebrand J, Heid<br />

IM; KORA, Lamina C, Gieger C, Illig T, Meitinger T, Wichmann HE, Herrera B, Hinney A, Hunt SE, Jarvelin<br />

MR, Johnson T, Jolley JD, Karpe F, Keniry A, Khaw KT, Luben RN, Mangino M, Marchini J, McArdle WL,<br />

McGinnis R, Meyre D, Munroe PB, Morris AD, Ness AR, Neville MJ, Nica AC, Ong KK, O'Rahilly S, Owen KR,<br />

Palmer CN, Papadakis K, Potter S, Pouta A, Qi L; Nurses' Health Study, Randall JC, Rayner NW, Ring SM,<br />

Sandhu MS, Scherag A, Sims MA, Song K, Soranzo N, Speliotes EK; Diabetes Genetics Initiative, Syddall HE,<br />

Teichmann SA, Timpson NJ, Tobias JH, Uda M; SardiNIA Study, Vogel CI, Wallace C, Waterworth DM,<br />

Weedon MN; Wellcome Trust Case Control Consortium, Willer CJ; FUSION, Wraight, Yuan X, Zeggini E,<br />

Hirschhorn JN, Strachan DP, Ouwehand WH, Caulfield MJ, Samani NJ, Frayling TM, Vollenweider P, Waeber<br />

G, Mooser V, Deloukas P, McCarthy MI, Wareham NJ, Barroso I, Jacobs KB, Chanock SJ, Hayes RB, Lamina<br />

C, Gieger C, Illig T, Meitinger T, Wichmann HE, Kraft P, Hankinson SE, Hunter DJ, Hu FB, Lyon HN, Voight<br />

BF, Ridderstrale M, Groop L, Scheet P, Sanna S, Abecasis GR, Albai G, Nagaraja R, Schlessinger D, Jackson<br />

AU, Tuomilehto J, Collins FS, Boehnke M, Mohlke KL. Common variants near MC4R are associated with fat<br />

mass, weight and risk of obesity. Nat Genet. 2008; 40: 768-75.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 44 of 74


MacDonald, S.L., Downing, I., Turner, M.L. & Kilpatrick, D.C. Is mannan-binding lectin (MBL) detectable on<br />

monocytes and monocyte-derived immature dendritic cells? Biochemical Society Transactions 2008; 36: 1497-<br />

1500<br />

McLintock LA, Cook G, Holyoake TL, Jones BL, Kinsey SE, Jackson GH. High loading dose AmBisome ® is<br />

efficacioius and well tolerated in the management of invasive fungal infection in haematology patients.<br />

Haematologica/The Hematology Journal. 2007: 92(04): 572-3.<br />

Mahmood, A., Gosling, P., Barclay, R., Kilvington, F., and Vohra, R. (2008). Splanchnic Microcirculation<br />

Protection by Hydroxyethyl Starches During Abdominal Aortic Aneurysm Surgery. Eur J Vasc Endovasc Surg.<br />

2009 ;37: 319-25.<br />

Mountford JC. Human embryonic stem cells: Origins, characteristics and potential for regenerative therapy.<br />

Transfus Med. 2008;18: 1-12.<br />

O’Looney N, Burgess STG, Chong Kwan M, Ross AJ, Robb JS, Forster T, Beattie JS, Ghazal P, Petrik J,<br />

Campbell C. Evaluation of a protein microarray method for immuno-typing erythrocytes in whole blood. Journal<br />

of Immunoassays & Immunochemistry, 2008; 29:197-209.<br />

Peden A, Head MW, Jones M, MacGregor I, Turner ML, Ironside J. Advances in the development of a<br />

screening test for vCJD. Expert Opinion on Medical Diagnostics 2008: 2: 207-219.<br />

Petrik J. Microbiological blood testing and new technologies. In “<strong>Transfusion</strong> Microbiology” (JAJ Barbara, MC<br />

Contreras and FAM Regan eds.) Cambridge University Press, 2008; 227-237.<br />

Prowse CV and Roberts D J. "<strong>Blood</strong> substitutes". Chapter 35 of "Practical <strong>Transfusion</strong> Medicine, Third<br />

edition" 2009 (editors Murphy MF , Pamphilon DH) pp390-399.<br />

Prowse CV. Pathogen inactivation of blood components. <strong>Transfusion</strong> Alternatives in <strong>Transfusion</strong> Medicine<br />

2008, 10, Issue 3, 139 -146 (& chapter II-E of planned NATA book).<br />

Prowse C. Pathogen Inactivated Plasma. <strong>Transfusion</strong> Medicine Reviews 2009; 23:124-33.<br />

Prowse C: Credit Accumulation and Transfer (CAT) & MSc Courses in <strong>Transfusion</strong> & Transplantation. BBTS<br />

<strong>Blood</strong>lines Spring 2008<br />

Prowse C. <strong>Blood</strong> Substitutes: Where are we now? <strong>Blood</strong> Matters (autumn 2008) 26: 21-24.<br />

Reesink HW, Engelfriet CP, Hyland CA, Coghlan P, Tait B, Wsolak M, Keller AJ, Henn G, Mayr WR, Thomas<br />

I, Osselaer JC, Lambermont M, Beaten M, Wendel S, Qiu Y, Georgsen J, Krusius T, Mäki T, Andreu G, Morel<br />

P, Lefrère JJ, Rebulla P, Giovanelli S, Butti B, Lecchi L, Mozzi F, van Hilten JA, Zwaginga JJ, Flanagan P,<br />

Flesland Ø, Brojer E, Letowska M, Akerblom O, Norda R, Prowse C, Dow B, Jarvis L, Davidson F, Kleinman<br />

S, Bianco C, Stramer SL, Dodd RY, Busch MP. Biobanks of blood from donors and recipients of blood<br />

products. Vox Sang. 2008 ; 94: 242-60.<br />

Rocchi MS, Anderson MJ, Eaton SL, Hamilton S, Finlayson J, Steele P, Barclay GR, Chianni F. Three colour<br />

flow cytometric detection of PrP in ovine leucocytes. Vet Immunol Immunopathol 2007; 116: 172-181.<br />

Shepherd P, Dhanapala C, Maguire C, White J, Drummond M, Holyoake T, Johnson PRE. Successful use of<br />

national cancer registry data to monitor the effective use of Imatinib for treating CML. Scott Med J. 2008;53:8-<br />

12<br />

Slight RD, Buell R, Nzewi OC, McClelland DB, Mankad PS. A comparison of activated coagulation time-based<br />

techniques for anticoagulation during cardiac surgery with cardiopulmonary bypass. J Cardiothorac Vasc<br />

Anesth. 2008 Feb;22(1):47-52<br />

Slight RD, O'Donohoe P, Fung AK, Alonzi C, McClelland DB, Mankad PS. Rationalizing blood transfusion in<br />

cardiac surgery: the impact of a red cell volume-based guideline on blood usage and clinical outcome. Vox<br />

Sang. 200;95:205-10<br />

Slight RD, Lux D, Nzewi OC, McClelland DB, Mankad PS. Oxygen delivery and hemoglobin concentration in<br />

cardiac surgery: when do we have enough? Artif Organs. 2008 Dec;32(12):949-55<br />

Slight RD, Alston RP, McClelland DB, Mankad PS. What factors should we consider in deciding when to<br />

transfuse patients undergoing elective cardiac surgery? Transfus Med Rev. 2009 Jan;23(1):42-54<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 45 of 74


Slight RD, Nzewi O, McClelland DB, Mankad PS. Red cell transfusion in elective cardiac surgery patients:<br />

where do we go from here?Br J Anaesth. 2009 Mar;102(3):294-296<br />

Slight RD, Ferguson R, Stirling D, McClelland DB, Mankad PS. Experience with sodium fluorescein flow<br />

cytometry in the determination of red cell volume Int J Lab Hematol. 2009 Apr;31(2):233-5<br />

Spence A, Townsend E, Prowse C, Palmer P, Fleming P, Joost A. Van Hilten, Ferguson E. A Multi-Level<br />

Analysis of the Stability of the Information Source-Trust Association for <strong>Blood</strong> <strong>Transfusion</strong>. <strong>Transfusion</strong> 2009:<br />

Apr 20. [Epub ahead of print]<br />

Swierzko AS, Atkinson APM, Cedzynski M, MacDonald SL, Szala A, Domzalska-Popadiuk I, Bokowska-Klos<br />

M, Jopek A, Szczapa J, Matsushita M, Szemraj J, Turner ML, Kilpatrick DC. Two factors of the lectin pathway<br />

of complement, L-ficolin and mannan-binding lectin, and their associations with prematurity, low birthweight<br />

and infections in a large cohort of Polish neonates. Mol Immunol. 2009;46:551-8<br />

Swierzko AS, Szala A, Cedzynski M, Domzalska-Popadiuk I, Borkowska-Klos M, Jopek A, Szczapa J,<br />

Szemraj J, Atkinson APM, MacDonalD SL, Turner ML, Kilpatrick DC. Mannan-binding lectin genotypes and<br />

genotype-phenotype relationships in a large cohort of Polish neonates. Hum Immunol. 2009;70:68-72<br />

Swierzko AS, Cedzynski M, Domzalska-Popadiuk I, MacDonald SL, Borkowska-Klos M, Atkinson AP, Szala A,<br />

Jopek A, Jensenius JC, Kawakami M, Szczapa J, Matsushita M, Szemraj J, Turner ML, Kilpatrick DC.<br />

Mannan-binding lectin-associated serine protease-2 (MASP-2) in a large cohort of neonates and its clinical<br />

associations Mol Immunol. 2009;46:1696-701<br />

Thomson RC, Petrik J, Nash AA, Dutia BM: Expansion and activation of NK cell populations in a<br />

gammaherpesvirus infection. Scandinavian J Immunology 2008; 67:489-95.<br />

Turner Ml. Transmission of vCJD by blood and tissues: managing the risk. The Biomedical Scientist 2007;<br />

Sept: 737-8.<br />

Turner ML, Hewitt P, Bruce M, Ironside JI. Prion Diseases. In: Barbara JAJ, Regan FAM, Contreras MC (eds).<br />

<strong>Transfusion</strong> Microbiology. 2 nd Edition. Cambridge University Press April 2008. 141-151.<br />

Turner ML. <strong>Transfusion</strong> Reactions. Chapter 75. In: Strachan MWJ, Sharma SK, Hunter JAA (eds). Davidson’s<br />

International Clinical Cases in Medicine. Churchill Livingstone, Elsevier. Edinburgh 2008.<br />

Turner ML. Variant CJD. In: Practical <strong>Transfusion</strong> Medicine. 3rd Edition Murphy MF, Pamphilion DH (eds).<br />

2008. Oxford: Blackwell Science.<br />

Turner ML. Prion Diseases. In: Rossi’s Principles of <strong>Transfusion</strong> Medicine 4th Edition, Simon TL, Snyder EL,<br />

Solheim BG, Stowell CP, Strauss RG, Petrides M (eds). Blackwell Publishing. Publisher: Wiley-Blackwell,<br />

ISBN:1405175885, 1112 pages, 2009).<br />

Turner ML, Ludlam CA. An Update on the Assessment and Management of the Risk of Transmission of<br />

variant Creutzfeldt-Jakob Disease by <strong>Blood</strong> and Plasma Products. Br J Haematol. 2009;144:14-23.<br />

Walsh TS "Perioperative blood component therapy and haemostasis" In: The Year in Anaesthesia and Critical<br />

Care. Eds. Hunter, Cook, Priebe, Struys. Clinical Publishing. Oxford 2008 (chapter)<br />

Ward FJ, Hall AM, Cairns LS, Leggat As, Urbaniak SJ, Vickers MA, Barker RN<br />

Clonal regulatory T cells specific for a red blood cell autoantigen in human autoimmune haemolytic anemia.<br />

<strong>Blood</strong> (2008) 111; 680-687<br />

Urbaniak SJ [editorial] Noninvasive approaches to the management of RhD haemolytic disease of the fetus<br />

and newborn. <strong>Transfusion</strong> (2008) 48; 2-5<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 46 of 74


Urbaniak, SJ. Why RHD is so immunogenic – and therefore important – and how we develop immune<br />

responses to blood cells. <strong>Blood</strong> Matters (2008), Issue 26, pp16-17<br />

<strong>SNBTS</strong> Active Grants 2008-09<br />

Wellcome Trust<br />

Turner ML, Crawford D, Haque T, Vickers MA, Forsythe J. Establishment of a bank<br />

of EBV specific cytotoxic T cells for clinical treatment of post-transplant lymphoproliferative<br />

disease. £431,069. April 2009 – Sept 2011.<br />

Turner ML, Mountford J, Forrester L, De Sousa P, Anstee D, Courtney A, Murphy<br />

W. Proof of principle: human embryonic stem cell derived red cell concentrates for<br />

clinical transfusion. £2,745,549 April 2009 – March 2012 (<strong>Blood</strong>Pharma)<br />

<strong>Scottish</strong> Funding Council.<br />

Wilmut I, Turner M, Illius A, Savill J. Strategic Development Grant for Centre of<br />

Regenerative Medicine. £2,030,000 (August 2006 to July 2010)<br />

UK Stem Cell Foundation<br />

Noble B. Autologous stem cell based therapies in musculoskeletal regenerative<br />

medicine. UK Stem Cell Foundation / Medical <strong>Research</strong> Council/ <strong>Scottish</strong><br />

Enterprise / Chief Scientist’s Office £1,400,000 May 2008 – May 2010.<br />

Dhillon B, Ramesh K, Turner ML. Ex vivo expanded corneal limbal stem cell<br />

transplantation: from laboratory to clinical application. UK Stem Cell Foundation,<br />

<strong>Scottish</strong> Enterprise and Chief Scientists Office. £307,820. Jan 2009 – Dec 2011<br />

Translational Medicine <strong>Research</strong> Collaboration<br />

Newby DE, Burdess A, Richards J, Lang N, Roddie H, McKillop G, Wardlaw J,<br />

Turner M, Hart S, Barclay R, Chalmers R, Simpson J, Dhaliwal K, Marshall I,<br />

Bastin M. Magnetic resonance imaging of human atherosclerosis. Translational<br />

Medicine <strong>Research</strong> Collaboration £566,995. June 2007 – August 2009<br />

Sir Jules Thorn Charitable Trust<br />

Forbes SJ, Iredale J, Hayes P, Kluth D, Turner ML, Bellamy C, Marshall I. Stem<br />

cell therapy for liver cirrhosis. £999,565. Oct 2007 – Sept 2012<br />

Medical <strong>Research</strong> Council<br />

Holyoake TL, Brunton V. Koschmieder S. Is Bcr-Abl expression relevant for the<br />

survival of cancer stem cells in chronic myeloid leukaemia? £680,000. (June 2007<br />

– May 2010)<br />

European Union<br />

SJ Urbaniak, SS Armstrong-Fisher, M Moss. Special non-invasive advances in<br />

fetal and neonatal evaluation network (SAFE). £113,195 (current share of 12<br />

million euro) SAFE Fellow & PhD student. EU (Network of Excellence) (2004-2009)<br />

Franklin IM, Hart M, Arthur E, McClelland DBL, Pirie E The EU optimal blood use<br />

project. 890,285 Euros €500,000 (56.16% EC funded) (3 years 2007-2010)<br />

Leukaemia <strong>Research</strong> Fund<br />

Holyoake TL. Brunton V, Koschmider S. Is Bcr-Abl expression relevant for the<br />

survival of cancer stem cells in chronic myeloid leukaemia? £28,221. (3 years<br />

January 2007 – December 2009)<br />

Holyoake TL, Bartholomew C, Strathdee G, Melo J. EVII isoform expression and<br />

knockdown in CML. Leukaemia <strong>Research</strong> Fund. £45,000. April 2008 – March<br />

2011.<br />

Biotechnology and Biological Sciences <strong>Research</strong> Council<br />

De Sousa P, Turner ML, Pells S. A novel characterisation and separation<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 47 of 74


technique for pluripotent human embryonic and haematopoietic stem cells. BBSRC<br />

£131,765. Jan 2009 – Dec 2012.<br />

British Heart Foundation<br />

Barclay GR, Hadoke P, Mills N, Newby DE, Turner ML. Preclinical in vivo<br />

evaluation of potential sources of human endothelial progenitor cells for autograft<br />

cellular therapy of ischaemia. £239,930 (3 years July 2006 to June 2009).<br />

Mills NL, Newby DE, Barclay GR, de Belder MA, Robinson SD. Endothelial<br />

progenitor cells in acute vascular injury and repair. £242,685 (August 2007-July<br />

2010)<br />

Mountford J. Generation of cardiomyocytes from mesenchymal stem cells derived<br />

from adult human sternal bone marrow. (as Principal Investigator for Clinical<br />

<strong>Research</strong> Fellow funding) £149,478 (3 years from January 2005-December 2008)<br />

Chief Scientist Office.<br />

De Sousa P, Head MW, Turner ML, Manson J. Proof of principle validation of a<br />

human embryo cell based screen for susceptibility to infectious prion transmission.<br />

£125,438. (August 2007 – December 2008).<br />

Head MW, Peden A, Prowse C, Manson J, Bishop M, Ironside J. The use of<br />

human recombinant prion protein as a substrate in an improved in vitro<br />

amplification- based blood test for Creutzfeldt-Jakob disease infectivity £193,512<br />

over 2 years to December 2010 (£90,000pa)<br />

Holyoake T, Copland MC. Consumables £24,000. (3 years June 2006 - May<br />

2009).<br />

De Sousa P, Head M, Turner M, Manson J. Proof of principle demonstration of<br />

human embryo stem cell based detection of prions.£127,707 (18 months to June<br />

2009).<br />

Walsh T, Forbes J, Langston A, McClelland DBL, Pelly J, Ramsay P, Watson D,<br />

Prowse C.<br />

A feasibility randomized trial comparing REstrictive and LIberal blood transfusion<br />

strategies in patients requiring six or more days in intEnsiVE care (RELIEVE study)<br />

£182,577 over 18 months to October 2010 (£100,000pa)<br />

Armstrong-Fischer SS, Urbaniak SJ. Non-Invasive Prenatal Fetal D typing (follow<br />

up on optimasation assay timing) £62, 259 over 12 months from June 2009<br />

Department of Health<br />

Houston F, Prowse C, Turner M, MacGregor I, Hornsey V, Hunter N, Foster J,<br />

Groschup M. The effect of leucodepletion on transmission of BSE by transfusion of<br />

sheep blood components. £3,424,382 (6 years to December 2010.)<br />

Extensionto 2011 and possible increase under negotiation by Jean Manson<br />

Navigant Inc.<br />

Prowse C et al. Validation of riboflavin treated fresh frozen plasma. £52,310 . (1<br />

year to July 2008)<br />

UK <strong>Blood</strong> Services Forum<br />

Prowse C, Turner M, Jones M, Head M: “ vCJD confirmatory assay developments”<br />

£179,563 over 3 years from 1 st April 2008-2011 (£60,000pa)<br />

<strong>SNBTS</strong> component of Prion filter trial (PRISM)<br />

<strong>National</strong> Services Scotland (<strong>Scottish</strong> <strong>National</strong> <strong>Blood</strong> <strong>Transfusion</strong> Service)<br />

Prowse C, Turner M, Jones M, Head M: for infrastructure and routine support of<br />

vCJD assay development CVP £65,000 capital and £80,648 pa salary and<br />

consumables over 3 years until 31st March 2011<br />

Polish State Committee for Scientific <strong>Research</strong><br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 48 of 74


Swierzko AS, Szczapa J, Cedzynski M, Szemraj J, Domzalska-Popadiuk, I, Klink<br />

M, Kilpatrick D.C. Lectin pathway of complement in defence against infection in<br />

newborns. £21,000 ( 3 years May 2005-May 2008).<br />

Cedzynski M, Atkinson APM, Swierzko AS, Szemraj J, MacDonald SL, Kilpatrick<br />

DC, Buczylko K, Zeman K. Investigation of the role of insufficiency of L-ficolin and<br />

other components of the lectin pathway of complement activation in recurrent<br />

respiratory infections in children with allergies. £26,000 (3 years April 2006- April<br />

2009).<br />

Iranian Government<br />

SJ Urbaniak, RN Barker, M Moss Gholamreza Sarab 2004-2008<br />

PhD studentship – HPA-1a GP IIIa peptide binding and processing.<br />

External overall funding = £93,900 . <strong>SNBTS</strong> contribution for 2008 – £5000<br />

consumables.<br />

SJ Urbaniak, SS Armstrong-Fisher. Ali Varzi 2004-2008<br />

PhD Studentship – identification of fetal DNA genetic markers in maternal plasma.<br />

£91,400. <strong>SNBTS</strong> contribution for 2008 – £5000 consumables.<br />

Saudi Arabian Government<br />

M Moss, S Urbaniak (Abdulla Meshi)<br />

Characterisation of the human humoral immune response to RhD derived synthetic<br />

peptides.<br />

£89,745 2008-2011<br />

DiaMed AG<br />

RH Fraser. Development of novel monoclonal blood grouping reagents (<strong>Research</strong><br />

Assistant salary funding). 200,000 SFr £88,000 (Rolling grant from years 2004-<br />

2008)<br />

NHS Grampian University Hospitals Division (GUHT) Endowments.<br />

SJ Urbaniak. <strong>Research</strong> Technician. (Sadie Henderson) 2007-2009<br />

£36,000.<br />

Nestech Partnership<br />

SJ Urbaniak, L Cairns, RN Barker, E Rattray. Peptide immunotherapy to suppress<br />

antibodies to red blood cells. £203,875. (3 years from 2005-2008)<br />

NHS <strong>National</strong> Services Scotland - Service Redesign Bid<br />

SJ Urbaniak, SS Armstrong-Fisher.<br />

Service Redesign Committee– pilot study of non-invasive RhD genotyping on<br />

maternal plasma. £56,480. 2006-2009.<br />

NHS <strong>Blood</strong> and Tissues (England)<br />

SS Armstrong-Fisher, SJ Urbaniak. Placental transfer of recombinant antibodies.<br />

£5000. <strong>SNBTS</strong> contribution for 2008 - £4000 2006-2008<br />

University of Bristol<br />

SS Armstrong-Fisher, SJ Urbaniak. Fetal DNA genetic markers in maternal<br />

plasma. (Funding support of PhD Studentship) £15,000 (in year 2007-2008)<br />

University of Bristol<br />

<strong>Scottish</strong> Enterprise Proof of Concept<br />

M Moss, S Urbaniak. <strong>Blood</strong> group mimotopes. £248,945. (3 years 2008-2010)<br />

Glasgow Royal Infirmary Endowments<br />

Holyoake TL and Allan E. Targeting quiescent CML stem cell by combining G-CSF<br />

and<br />

imatinib mesylate £9,700 (6 years to January 2011)<br />

Holyoake T, Mysinna S. Is survival of primitive, quiescent stem cells from patients<br />

with chronic myeloid leukaemia Bcr-Abl dependent or not?<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 49 of 74


£31,000. January 2008 – November 2008<br />

Alba BioScience<br />

Juraj Petrik Microarray diagnostics for blood borne viruses<br />

2009:£ ~ 56,000. 2010:£ ~ 58,000<br />

2009-2010<br />

Allan EK, Hamilton A, Hatziieremia S, Zhou P, Jorgensen HG, Vigneri P, Holyoake TL. Nuclear<br />

entrapment of BCR-ABL by combining imatinib mesylate with leptomycin B does not eliminate CD34+<br />

chronic myeloid leukaemia cells. Leukemia. 2009; 23: 1006-8<br />

Anani Sarab G, Moss M, Barker RN, Urbaniak SJ. Naturally processed peptides spanning the HPA-1a<br />

polymorphism are efficiently generated and displayed from platelet glycoprotein by HLA-DRB3*0101 -<br />

positive antigen-presenting cells. <strong>Blood</strong>. 2009;114:1954-7.<br />

Ban M, Goris A, Lorentzen AR, Baker A, Mihalova T, Ingram G, Booth DR, Heard RN, Stewart GJ,<br />

Bogaert E, Dubois B, Harbo HF, Celius EG, Spurkland A, Strange R, Hawkins C, Robertson NP,<br />

Dudbridge F, Wason J, De Jager PL, Hafler D, Rioux JD, Ivinson AJ, McCauley JL, Pericak-Vance M,<br />

Oksenberg JR, Hauser SL, Sexton D, Haines J, Sawcer S; Wellcome Trust Case-Control Consortium<br />

(WTCCC), Compston A. Replication analysis identifies TYK2 as a multiple sclerosis susceptibility factor.<br />

Eur J Hum Genet. 2009; 17: 1309-13.<br />

Bellodi C, Lidonnici MR, Hamilton A, Helgason GV, Soliera AR, Ronchetti M, Galavotti S, Young KW,<br />

Selmi T, Yacobi R, Van Etten RA, Donato N, Hunter A, Dinsdale D, Tirrò E, Vigneri P, Nicotera P, Dyer<br />

MJ, Holyoake T, Salomoni P, Calabretta B. Targeting autophagy potentiates tyrosine kinase inhibitorinduced<br />

cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin<br />

Invest. 2009 May;119(5):1109-23.<br />

Bessos H, Killie MK, Seghatchian J, Skogen B, Urbaniak SJ. The relationship of anti-HPA-1a amount to<br />

severity of neonatal alloimmune thrombocytopenia - Where does it stand? Transfus Apher Sci.<br />

2009;40:75-8.<br />

Biggin K, Warner P, Prescott R, McClelland B. A review of methods used in comprehensive, descriptive<br />

studies that relate red blood cell transfusion to clinical data. <strong>Transfusion</strong>. 2010; 50: 711-718.<br />

British Committee for Standards in Haematology (Harris AM (BCSH Lead), Atterbury CLJ, Chaffe B,<br />

Elliott C, Hawkins T, Hennem SJ, Howell C, Jones J, Murray S, New HV, Norfolk D, Pirie L (British<br />

Transplantation Society Lead), Russell J, Taylor C. Guideline on the Administration of <strong>Blood</strong><br />

Components. London: British Society for Haematology. BCSH web site. 2009; December: 1-60.<br />

Cedzynski M, Atkinson APM, Swierzko AS, MacDonald SL, Szala A, Zeman K, Buczylko K, Bak-<br />

Romaniszyn L, Wiszniewska M, Matsushita M, Szemraj J, Banasik M, Turner ML, Kilpatrick DC. L-Ficolin<br />

(Ficolin-2) insufficiency is associated with combined allergic and infectious respiratory disease in<br />

children. Molecular Immunology 2009; 47: 415-419.<br />

Chapman CE, Stainsby D, Jones H, Love E, Massey E, Win N, Navarrete C, Lucas G, Soni N, Morgan<br />

C, Choo L, Cohen H, Williamson LM; on behalf of the Serious Hazards of <strong>Transfusion</strong> Steering Group<br />

(McClelland DBL et al). Ten years of hemovigilance reports of transfusion-related acute lung injury in the<br />

United Kingdom and the impact of preferential use of male donor plasma. <strong>Transfusion</strong>. 2009; 49: 440-<br />

452.<br />

Clark P, Wu O, Greer IA, Lowe GDO. Venous thrombosis prevention – more that just guidelines. British<br />

Journal of Haematology. 2010; 149: 50-54<br />

Coste J, Prowse C, Eglin R, Fang C. Subgroup on TSE. A report on transmissible spongiform<br />

encephalopathies and transfusion safety. Vox Sang. 2009: 96: 284-291.<br />

Crawford J, Turner M. Stem cell therapies: hype or reality? J R Coll Physicians Edinb 2008; 38: 221-3<br />

Davies A, Jordanides NE, Giannoudis A, Lucas CM, Hatziieremia S, Harris RJ, Jørgensen HG, Holyoake<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 50 of 74


TL, Pirmohamed M, Clark RE, Mountford JC. Nilotinib concentration in cell lines and primary CD34(+)<br />

chronic myeloid leukemia cells is not mediated by active uptake or efflux by major drug transporters.<br />

Leukemia. 2009; 23: 1999-2006.<br />

Davidson F, Yirrell, D, Lycett C, Patrik J, Dow BC. HIV subtypes detected in <strong>Scottish</strong> blood donors. Vox<br />

Sang. 2009; 96: 160-162.<br />

Davison KL, Dow B, Barbara JA, Hewitt PE, Eglin R. The introduction of anti-HTLV testing of blood<br />

donations and the risk of transfusion-transmitted HTLV, UK: 2002-2006. Transfus Med. 2009;19:24-34<br />

Dehghan A, Yang Q, Peters A, Basu S, Bis JC, Rudnicka AR, Kavousi M, Chen MH, Baumert J, Lowe<br />

GD, McKnight B, Tang W, de Maat M, Larson MG, Eyhermendy S, McCardle WL, Lumley T, Pankow JS,<br />

Hofman A. Massaro JM, Rivadeneira F, Kolz M, Taylor KD, van Dujin CM, Kathiresan S, Illig T,<br />

Aulchenko YS, Volcik KA, Johnson AD, Uitterlinden AG, Tofler GH, Gieger C, Wellcome Trust Case<br />

Control Consortium, Psaty BM, Couper DJ, Boerwinkle E, Koenig W, O’Donnell CJ, Witteman JC,<br />

Strachan DP, Smith NL, Folsom AR. Association of novel genetic Loci with circulating fibrinogen levels:<br />

a genome-wide association study in 6 population-based cohorts. Circ Cardiovasc Genet. 2009; 2(2):<br />

125-133<br />

Drummond MW, Heaney N, Kaeda J, Nicolini FE, Clark RE, Wilson G, Shepherd P, Tighe J, McLintock<br />

L, Hughes T, Holyoake TL.A pilot study of continuous imatinib vs pulsed imatinib with or without G-CSF<br />

in CML patients who have achieved a complete cytogenetic response. Leukemia. 2009; 23:1199-201.<br />

Ferguson E, Spence A, Townsend E, Prowse C, Palmer J, Fleming P, Van Hilten JA (RASPH <strong>Research</strong><br />

Group) What type of information is trusted by whom? A multi-level analysis of the stability of the<br />

information source-trust association for blood transfusion. <strong>Transfusion</strong>. 2009; Apr 20. [Epub ahead of<br />

print]<br />

Fielding AK, Rowe JM, Richards SM, Buck G, Moorman AV, Durrant IJ, Marks DI, McMillan AK, Litzow<br />

MR, Lazarus HM, Foroni L, Dewald G, Franklin IM, Luger SM, Paietta E, Wiernik PH, Tallman MS,<br />

Goldstone AH. Prospective outcome data on 267 unselected adult patients with Philadelphia<br />

chromosome-positive acute lymphoblastic leukemia confirms superiority of allogeneic transplantation<br />

over chemotherapy in the pre-imatinib era: results from the International ALL Trial MRC<br />

UKALLXII/ECOG2993. <strong>Blood</strong>. 2009 ;113:4489-96.<br />

Foo J, Drummond MW, Clarkson B, Holyoake T, Michor F. Eradication of chronic myeloid leukemia stem<br />

cells: a novel mathematical model predicts no therapeutic benefit of adding G-CSF to imatinib. PLoS<br />

Comput Biol. 2009 Sep;5(9):e1000503. Epub 2009 Sep 11.<br />

Fotaki V, Larralde O, Zeng S, McLaughlin D, Nichols J, Price DJ, Theil T, Mason JO. Loss of Wnt8b has<br />

no overt effect on hippocampus development but leads to altered Wnt gene expression levels in<br />

dorsomedial telencephalon. Development Dynamics. 2010; 239: 284-296.<br />

Goldman JM, Green AR, Holyoake T, Jamieson C, Mesa R, Mughal T, Pellicano F, Perrotti D, Skoda R,<br />

Vannucchi AM.Chronic myeloproliferative diseases with and without the Ph chromosome: some<br />

unresolved issues. Leukemia. 2009; 23: 1708-15.<br />

Hamilton A, Alhashimi F, Myssina S, Jorgensen HG, Holyoake TL.Optimization of methods for the<br />

detection of BCR-ABL activity in Philadelphia-positive cells. Exp Hematol. 2009; 37: 395-401.<br />

Harrison SJ, Franklin IM, Campbell JD. Enumeration of blood dendritic cells in patients with multiple<br />

myeloma at presentation and through therapy. Leuk Lymphoma. 2008; 49: 2272-83.<br />

Hatziieremia S, Jordanides NE, Holyoake TL, Mountford JC, Jørgensen HG. Inhibition of MDR1 does not<br />

sensitize primitive chronic myeloid leukemia CD34+ cells to imatinib. Exp Hematol. 2009;37:692-700.<br />

Hochhaus A, O'Brien SG, Guilhot F, Druker BJ, Branford S, Foroni L, Goldman JM, Müller MC, Radich<br />

JP, Rudoltz M, Mone M, Gathmann I, Hughes TP, Larson RA; IRIS Investigators. Six-year follow-up of<br />

patients receiving imatinib for the first-line treatment of chronic myeloid leukemia. Leukemia. 2009; 23:<br />

1054-61.<br />

Holotnakova T, Tylkova L, Takacova M, Kopacek J, Petrik J, Pastorekova S, Pastorek J. Role of the HBx<br />

oncoprotein in carbonic anhydrase 9 induction. J Med Virol 2009; 82:32-40<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 51 of 74


Hornsey VS, Drummond O, Morrison A, McMillan L, MacGregor IR, Prowse CV. Pathogen reduction of<br />

fresh plasma using riboflavin and ultraviolet light: effects on plasma coagulation proteins. <strong>Transfusion</strong>.<br />

2009; 49: 2167-2172.<br />

Jones M, Peden AH, Yull H, Wight D, Bishop MT, Prowse CV, Turner ML, Ironside JW, MacGregor IR,<br />

Head MW. Human platelets as a substrate source for the in vitro amplification of the abnormal prion<br />

protein (PrP) associated with variant Creutzfeldt-Jakob disease. <strong>Transfusion</strong>. 2009; 49: 376-84<br />

Jones M, Wight D, Barron R, Jeffrey M, Manson J, Prowse C, Ironside JW, Head MW. Molecular model<br />

of prion transmission to humans. Emerging Infectious Diseases. 2009; 15: 2013-2016.<br />

Jones M, Wight d, Barron R, Jeffrey M, Prowse C, Ironside JW, Head MW. Modeling the Molecular<br />

component of Prion Transmission to Humans. In preparation for Nature Medicine 2009 .<br />

Jones M, Wight D, McLoughlin V, Norrby K, Ironside JW, Connolly JG, Farquhar CF, MacGregor IR,<br />

Head MW. An antibody to the aggregated synthetic prion protein peptide (PrP 106-126) selectively<br />

recognizes disease-associated prion protein (PrP) from human brain specimens. Brain Pathol. 2009;<br />

19: 293-302.<br />

Jones M, McLoughlin V, Connolly JG, Farquhar CF, MacGregor IR, Head MW. Production and<br />

characterization of a panel of monoclonal antibodies against native human cellular prion protein.<br />

Hybridoma (Larchmt). 2009; 28: 13-20.<br />

Joseph BG, Hendry C, Walsh TS. Red blood cell use outside the operating theater: a prospective<br />

observational study with modeling of potential blood conservation during severe blood shortages.<br />

<strong>Transfusion</strong>. 2009; 49: 2060-2069.<br />

Kilpatrick DC, Chalmers JD, MacDonald SL, Murray MP, Mohammed A, Hart SP, Matsushita M, Hill AT.<br />

Stable bronchiectasis is associated with low serum L-ficolin concentrations. Clinical Respiratory Journal<br />

2009, 3, 29-33<br />

Mahmood A, Gosling P, Barclay R, Kilvington F, Vohra R. Splanchnic microcirculation protection by<br />

hydrozyethyl starches during abdominal aortic aneurysm surgery. Eur. J. Vasc. Endovasc. Surg. 2009;<br />

37: 319-325.<br />

Milojkovic D, Nicholson E, Apperley JF, Holyoake TL, Shepherd P, Drummond MW, Szydlo R, Bua M,<br />

Foroni L, Reid A, Khorashad JS, de Lavallade H, Rezvani K, Paliompeis C, Goldman JM, Marin D. Early<br />

prediction of success or failure using second generation tyrosine kinase inhibitors for chronic myeloid<br />

leukemia. Haematologica. 2009 Oct 14. [Epub ahead of print]<br />

Mills NL, Tura O, Padfield GJ, Millar C, Lang NN, Stirling D, Ludlam C, Turner ML, Barclay GR, Newby<br />

DE. Dissociation of phenotypic and functional endothelial progenitor cells in patients undergoing<br />

percutaneous coronary intervention. Heart. 2009; 95: 2003-2008.<br />

Mountford J, Olivier E, Turner M. Prospects for the manufacture of red cells for transfusion. British<br />

Journal of Haematology. 2010; 149: 22-34.<br />

Myssina S, Helgason GV, Serrels A, Jørgensen HG, Bhatia R, Modi H, Baird JW, Mountford JC,<br />

Hamilton A, Schemionek M, Koschmieder S, Brunton VG, Holyoake TL. Combined BCR-ABL inhibition<br />

with lentiviral-delivered shRNA and dasatinib augments induction of apoptosis in Philadelphia-positive<br />

cells. Exp Hematol. 2009; 37: 206-14.<br />

Nicholson E, Holyoake T. The chronic myeloid leukaemia stem cell. Clin. Lymphoma Myeloma. 2009;<br />

9: Suppl. 4: S376-81 Review.<br />

Orozco G, Hinks A, Eyre S, Ke X, Gibbons LJ, Bowes J, Flynn E, Martin P, Wellcome Trust Case Control<br />

Consortium, YEAR consortium, Wilson AG, Bax DE, Morgan AW, Emery P, Steer S, Hocking L, Reid<br />

DM, Wordsworth P, Harrison P, Thomson W, Barton A, Worthington J. Combined effects of three<br />

independent SNPs greatly increase the risk estimate for RA at 6q23. Hum Mol Genet. 2009; 18: 2693-<br />

2699.<br />

Pellicano F, Copland M, Jorgensen HG, Mountford J, Leber B, Holyoake TL. BMS-214662 induces<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 52 of 74


mitochondrial apoptosis in chronic myeloid leukemia (CML) stem/progenitor cells, including CD34+38-<br />

cells, through activation of protein kinase Cbeta. <strong>Blood</strong>. 2009;114:4186-96<br />

Pellicano F, Cilloni D, Helgason GV, Messa F, Panuzzo C, Arruga F, Bracco E, Allan E, Huntly BJ,<br />

Holyoake TL, Saglio G. FOXO transcription factor activity is partially retained in quiescent CML stem<br />

cells and induced by tyrosine kinase inhibitors in CML progenitor cells. <strong>Blood</strong>. 2009 Dec 1. [Epub ahead<br />

of print]<br />

Petrik J. Microarray blood testing: Pros & cons. Biologicals. 2010; 38: 2-8.<br />

Pirie, L. Green, J. (2009) A framework to support Nurses and Midwives making the clinical decision and<br />

providing the written instruction for blood component transfusion.<br />

http://www.transfusionguidelines.org.uk/Index.aspx?Publication=BBT&Section=22&pageid=1298<br />

Prowse C. Properties of pathogen-inactivated plasma components. Transfus. Med. Rev. 2009; 23:<br />

124-133.<br />

Reesink HW, Panzer S, McQuilten ZK, Wood EM, Marks DC, Wendel S, Trigo F, Biagini S, Olyntho S,<br />

Devine DV, Mumford I, Cazenave JP, Rasonglès P, Garraud O, Richard P, Schooneman F, Vezon G, Al<br />

Radwan R, Brand A, Hervig T, Castro E, Lozano M, Navarro L, Puig L, Almazán C, Maclennan S,<br />

Cardigan R, Franklin IM, Prowse C. Pathogen inactivation of platelet concentrates. Vox Sang. 2010 Mar<br />

10. [Epub ahead of print]<br />

Reikvam H, Prowse C, Roddie H, Heddle NM, Hervig T (for the BEST collaborative). A pilot study of the<br />

possibility and the feasibility of haemoglobin dosing with red blood cells transfusion. Vox Sanguinis.<br />

2010; Mar 10 [Epub ahead of print]<br />

Robb AO, Mills NL, Smith IB, Short A, Tura-Ceide O, Barclay GR, Blomberg A, Critchley HO, Newby DE,<br />

Denison FC. Influence of menstrual cycle on circulating endotheilial progenitor cells. Hum. Reprod.<br />

2009; 24: 619-625.<br />

Schemionek M, Elling C, Steidl U, Bäumer N, Hamilton A, Spieker T, Göthert JR, Stehling M, Wagers A,<br />

Huettner CS, Tenen DG, Tickenbrock1 L, Berde WE, Hubert Serve H, Holyoake TL, Müller-Tidow C,<br />

Koschmieder S. BCR-ABL enhances differentiation of long-term repopulating hematopoietic stem cells<br />

<strong>Blood</strong> 2010; 115: 3185-3195.<br />

Slight RD, Alston RP, McClelland DB, Mankad PS. What factors should we consider in deciding when to<br />

transfuse patients undergoing elective cardiac surgery? Transfus Med Rev. 2009;23:42-54.<br />

Sullivan, G. J., Hay, D. C., Park, I. H., Fletcher, J., Hannoun, Z., Payne, C. M., Dalgetty, D., Black, J. R.,<br />

Ross, J. A., Samuel, K., Wang, G., Daley, G. Q., Lee, J. H., Church, G. M., Forbes, S. J., Iredale, J. P.,<br />

and Wilmut, I. Generation of functional human hepatic endoderm from human induced pluripotent stem<br />

cells. Hepatology 2009, (Epub ahead of print)<br />

Swierzko AS, Atkinson APM, Cedzynski M, MacDonald SL, Szala A, Domzalska-Popadiuk I, Bokowska-<br />

Klos M, Jopek A, Szczapa J, Matsushita M, Szemraj J, Turner ML, Kilpatrick DC. Two factors of the<br />

lectin pathway of complement, L-ficolin and mannan-binding lectin, and their associations with<br />

prematurity, low birthweight and infections in a large cohort of Polish neonates. Molecular Immunology<br />

2009; 46: 551-558.<br />

Swierzko, AS, Szala A, Cedzynski M, Domzalska-Popadiuk I, Borkowska-Klos M, Jopek A, Szczapa J,<br />

Szemraj J, Atkinson APM, MacDonald SL, Turner ML, Kilpatrick DC. Mannan-binding lectin genotypes<br />

and genotype-phenotype relationships in a large cohort of Polish neonates. Human Immunology 2009;<br />

70: 68-72.<br />

Swierzko AS, Cedzynski M, Domzalska-Popadiuk I, MacDonald SL, Borkowska-Klos M, Atkinson APM,<br />

Szala A, Jopek A, Jensenius J, Kawakami M, Sxcxapa J, Matsushita M, Szemraj J, Turner ML, Kilpatrick<br />

DC. Mannan-binding lectin associated serine protease-2 (MASP-2) in a large cohort of neonates and its<br />

clinical associations. Molecular Immunology 2009; 46: 1696-1701.<br />

Szabo SM, Levy AR, Davis C, Holyoake TL, Cortes J. A Multinational Study of Health State Preference<br />

Values Associated with Chronic Myelogenous Leukemia. Value Health. 2009 Jul 29. [Epub ahead of<br />

print]<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 53 of 74


Taylor K, Rolfe M, Reynolds N, Kilanowski F, Pathania U, Clarke D, Yang D, Oppenheim J, Samual K,<br />

Howie S, Barran P, Macmillan D, Campopiano D, Dorin J. Defensin-related peptide 1 (Defr 1) is allelic to<br />

Defb8 and chemoattracts immature DC and CD4+T cells independently of CCR6. Eur. J. Immunol.<br />

2009; 39: 1353-1360.<br />

Terrace JD, Hay DC, Samuel K, Payne C, Anderson RA, Currie IS, Parks RW, Forbes SJ, Ross JA.<br />

Side population cells in developing human liver are primarily haematopoietic progenitor cells. Exp. Cell.<br />

Res. 2009; 315: 2141-2153.<br />

Turner ML. Prion diseases. In: Rossi’s Principles of <strong>Transfusion</strong> Medicine 4th Edition, Simon TL, Snydr<br />

EL, Solheim BG, Stowell CP, Strauss RG, Petrides M (eds). 2009 Blackwell Publishing 791-800.<br />

Urbaniak SJ. Alloimmunization to the platelet antigen human platelet antigen-1a and characterization of<br />

the helper T-cell responses in Fetomaternal Alloimmune thrombocytopenia. Vox Sang. 2009; 41)<br />

107-110.<br />

Urbaniak SJ. Recommendations of the ISBT working party on granulocytes immunobiology for<br />

leucocyte antibody screening in the investigation and prevention of antibody-mediated transfusionrelated<br />

acute lung injury ISBT working party on granulocytes immunobiology. Vox Sang. 2009; 96: (3)<br />

266-269.<br />

Vanhoutte VJ, McAulay KA, McCarrell E, Turner M, Crawford DH, Haque T. Cytolytic mechanisms and<br />

T-cell receptor Vbeta usage by ex vivo generated Epstein-Barr virus-specific cytotoxic T lymphocytes.<br />

Immunology. 2009; 127: 577-586.<br />

Walsh TS, MacIver CR. A clinical scenario-based survey of transfusion decisions for intensive care<br />

patients with delayed weaning from mechanical ventilation. <strong>Transfusion</strong>. 2009; 49: 2661-2667.<br />

Walsh TS, MacIver for The <strong>Scottish</strong> Critical Care Trials Group And <strong>Scottish</strong> <strong>National</strong> <strong>Blood</strong> <strong>Transfusion</strong><br />

Service Clinical Effectiveness Group. A clinical scenario-based survey of transfusion decisions for<br />

intensive care patients with delayed weaning from mechanical ventilation. <strong>Transfusion</strong>. 2009 Aug 4.<br />

[Epub ahead of print]<br />

Franklin IM. Introduction: recent evolution of transfusion medicine. In: Practical <strong>Transfusion</strong> Medicine<br />

(3rd Edition). Murphy MF, Pamphilon DH (Eds). Oxford: Blackwell Publishing. 2009; Ch1: 1-4<br />

McClelland B, Walsh T. Good blood management: the effective and safe use of blood components. In:<br />

Practical <strong>Transfusion</strong> Medicine (3rd Edition). Murphy MF, Pamphilon DH (Eds). Oxford: Blackwell<br />

Publishing. 2009; Ch 25: 265-284.<br />

McQuaker IG, Franklin IM. Haemopoietic stem cell transplantation. In: Practical <strong>Transfusion</strong> Medicine<br />

(3rd Edition). Murphy MF, Pamphilon DH (Eds). Oxford: Blackwell Publishing. 2009; Ch 40: 443-452.<br />

Prowse CV and Roberts D J. <strong>Blood</strong> substitutes. In: Practical <strong>Transfusion</strong> Medicine (3rd Edition). Murphy<br />

MF, Pamphilon DH (Eds). Oxford: Blackwell Publishing. 2009; Ch 35: 390-399.<br />

Turner ML. Variant Creutzfeldt-Jakob disease. In: Practical <strong>Transfusion</strong> Medicine (3rd Edition). Murphy<br />

MF, Pamphilon DH (Eds). Oxford: Blackwell Publishing. 2009; Ch 15: 153-161.<br />

Turner ML. Prion Diseases. In: Rossi’s Principles of <strong>Transfusion</strong> Medicine 4th Edition, Simon TL, Snyder<br />

EL, Solheim BG, Stowell CP, Strauss RG, Petrides M (eds). 2009 Blackwell Publishing. 791-800.<br />

Aubuchon J, Prowse C. AABB monograph on Pathogen Inactivation – in preparation 2009 (Chapters<br />

and book editor)<br />

Dow BC, Franklin IM, Munro H, Gunson R. Syphilis nucleic acid testing: usefulness in syphilis<br />

confirmation? <strong>Transfusion</strong>. 2010; 50(3): 737-739.<br />

Franklin IM. Prevention of rhesus haemolytic disease of the fetus and newborn. Lancet. 2009;373:1082.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 54 of 74


<strong>SNBTS</strong> Active Grants 2009-10<br />

Wellcome Trust<br />

Turner ML, Crawford<br />

D, Haque T, Vickers<br />

MA, Forsythe J.<br />

Turner ML, Mountford<br />

J, Forrester L, De<br />

Sousa P, Anstee D,<br />

Courtney A, Murphy<br />

W.<br />

Establishment of a bank of EBV<br />

specific cytotoxic T cells for clinical<br />

treatment of post-transplant<br />

lympho-proliferative disease.<br />

Proof of principle: human<br />

embryonic stem cell derived red<br />

cell concentrates for clinical<br />

transfusion.<br />

[‘<strong>Blood</strong> Pharma’]<br />

£ 431,069<br />

(. Jan 2010 – June 2012)<br />

£2,745,549<br />

(3 years: July 2009 – June<br />

2012)<br />

<strong>Scottish</strong> Funding Council<br />

Wilmut I, Turner M,<br />

Illius A, Savill J<br />

Strategic Development Grant for<br />

Centre of Regenerative Medicine<br />

2,030,000<br />

(4 years August 2006 to<br />

July 2010)<br />

UK Stem Cell Foundation/ Medical <strong>Research</strong> Council/<strong>Scottish</strong> Enterprise/Chief Scientist’s Office<br />

Noble B, Turner ML. Autologous stem cell based<br />

therapies in musculoskeletal<br />

regenerative medicine.<br />

£1,400,000<br />

(2 years May 2008 to May<br />

2010)<br />

CZB/4/6<br />

57<br />

Dhillon B, Ramaesh K,<br />

Turner ML.<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£307,820<br />

(3 years January 2009 to<br />

December 2011)<br />

Translational Medicine <strong>Research</strong> Collaboration<br />

Newby DE, Burdess<br />

A, Richards J, Lang N,<br />

Roddie H, McKillop G,<br />

Wardlaw J, Turner M,<br />

Hart S, Barclay R,<br />

Chalmers R, Simpson<br />

J, Dhaliwal K, Marshall<br />

I, Bastin M.<br />

Magnetic resonance imaging of<br />

human atherosclerosis.<br />

£566,995<br />

(2 years June 2007 to<br />

August 2009)<br />

Sir Jules Thorn Charitable Trust<br />

Forbes SJ, Iredale J,<br />

Hayes P, Kluth D,<br />

Turner ML, Bellamy C,<br />

Marshall I.<br />

Stem cell therapy for liver cirrhosis £999,565<br />

(5 years October 2007 to<br />

September 2012)<br />

Medical <strong>Research</strong> Council<br />

Holyoake TL, Brunton<br />

V. Koschmieder S.<br />

Holyoake T,<br />

Koschmieder S,<br />

Calabretta B, Salmoni<br />

P.<br />

Is Bcr-Abl expression relevant for<br />

the survival of cancer stem cells in<br />

chronic myeloid leukaemia?<br />

Is the introduction of autophagy by<br />

tyrosine kinase inhibitors a key<br />

survival mechanism for chromic<br />

myeloid leukaemia stem cells?<br />

£680,000<br />

(3 years June 2007 to May<br />

2010)<br />

£1,117,312. Proposed start<br />

date 1st January 2010 for 3<br />

years.<br />

European Union – Network of Excellence<br />

Urbaniak SJ,<br />

Armstrong-Fisher SS,<br />

Moss M.<br />

Special non-invasive Advances in<br />

Foetal and Neonatal Evaluation<br />

network (SAFE) on non-invasive<br />

foetal RhD genotyping and<br />

evaluation<br />

(SAFE Fellow & PhD studentship)<br />

£113,195<br />

Share of 12m Euros (over 5<br />

years 2004 to 2009 with 19<br />

core laboratories)<br />

2004 – 2009<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 55 of 74


European Union – Public Health Executive Agency<br />

Franklin IM, Hart M, EU Optimal <strong>Blood</strong> Use Project<br />

Arthur E, McClelland<br />

DBL, Pirie E<br />

€890,285 Euros<br />

€500,000 (56.16% EC<br />

funded)<br />

(3 years 2007 – 2010)<br />

Leukaemia <strong>Research</strong> Fund<br />

LRF<br />

Grant No<br />

06075<br />

Holyoake TL. Brunton<br />

V, Koschmider S<br />

Is Bcr-Abl expression relevant for<br />

the survival of cancer stem cells in<br />

chronic myeloid leukaemia?<br />

£28,221 (3 years Oct 2007 -<br />

Sept 2010.)<br />

LRF<br />

Grant No<br />

08018<br />

Holyoake TL,<br />

Bartholomew C,<br />

Strathdee G, Melo J.<br />

EVII isoform expression and<br />

knockdown in CML.<br />

£45,000<br />

(3 years April 2008 to March<br />

2011)<br />

LRF<br />

Grant No<br />

08071<br />

Whetton T,<br />

Holyoake T. A<br />

phosphoproteomic investigation of<br />

key BCR-ABL mediated signally<br />

pathways in CML stem cells: where<br />

does tyrosine kinase inhibitor<br />

resistance lie?.<br />

£353,479 Jan 09 – Dec 2011<br />

Cancer <strong>Research</strong> United Kingdom.<br />

Holyoake T, Whetton<br />

T, Girolami M.<br />

Key survival pathways in chronic<br />

myeloid leukaemia (CML) stem<br />

cells and novel approaches to their<br />

eradication. Cancer <strong>Research</strong><br />

United Kingdom.<br />

Biotechnology, Biophysical Scientific <strong>Research</strong> Council (BBSRC)<br />

De Sousa P, Turner<br />

ML, Pells S.<br />

A novel characterisation and<br />

separation technique for pluripotent<br />

human embryonic and<br />

haematopoietic stem cells.<br />

£1.1 million. Proposed start<br />

date 1st January 2010 2009<br />

for 5 years. Amount applied<br />

for £1,138,122.<br />

£131,765<br />

(3 years January 2009 to<br />

December 2012)<br />

British Heart Foundation<br />

BHF<br />

Project<br />

Grant No<br />

PG/06/0<br />

51<br />

BHF<br />

Project<br />

Grant No<br />

PG/07/0<br />

17/2240<br />

5<br />

Barclay GR, Mills N,<br />

Newby D, Hadoke P,<br />

Turner ML.<br />

Mills NL, Newby DE,<br />

Barclay GR, de Belder<br />

MA, Robinson SD<br />

Preclinical in vivo evaluation of<br />

potential sources of human<br />

endothelial progenitor cells for<br />

autograft cellular therapy of<br />

ischaemia<br />

Endothelial progenitor cells in<br />

acute vascular injury and repair.<br />

£239,930<br />

(3 years July 2006 to June<br />

2009)<br />

- extended to end June<br />

2010 (i.e. 4 years total)<br />

because of 2 sets of<br />

maternity leave by O Tura).<br />

£242,685<br />

(3 years August 2007 to July<br />

2010)<br />

Department of Health<br />

Manson J, Houston F,<br />

Prowse C, Turner M,<br />

MacGregor I, Hornsey<br />

V, Hunter N,<br />

Foster J, Groschup M.<br />

The effect of leucodepletion on<br />

transmission of BSE by transfusion<br />

of sheep blood components.<br />

£3,424,382 (6 years to<br />

December 2010)<br />

Extension to 2012 agreed<br />

with DoH & Jean Manson<br />

Chief Scientist Office – <strong>Scottish</strong> Executive Health Department (CSO SEHD)<br />

Holyoake T, Copland<br />

MC<br />

Consumables 24,000. (3 years June 2006<br />

to May 2009.<br />

De Sousa P, Head .. Proof of principle validation of a £125,438. Aug 2007 – Dec<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 56 of 74


MW, Turner ML,<br />

Manson J<br />

Head MW, Peden A,<br />

Prowse C, Manson J,<br />

Bishop M, Ironside J.<br />

Walsh T, Forbes J,<br />

Langston A,<br />

McClelland DBL, Pelly<br />

J, Ramsay P, Watson<br />

D, Prowse C.<br />

human embryo cell based screen<br />

for susceptibility to infectious prion<br />

transmission.<br />

The use of human recombinant<br />

prion protein as a substrate in an<br />

improved in vitro amplificationbased<br />

blood test for Creutzfeldt-<br />

Jakob disease infectivity<br />

A feasibility randomized trial<br />

comparing REstrictive and LIberal<br />

blood transfusion strategies in<br />

patients requiring six or more days<br />

in intEnsiVE care (RELIEVE study)<br />

2009.<br />

£193,512 over 2 years to<br />

December 2010<br />

(£90,000pa)<br />

£182,577 over 18 months to<br />

October 2010<br />

(£100,000pa)<br />

Grant No<br />

CAF/08/<br />

09.<br />

Grant no<br />

CZB/4/6<br />

90.<br />

CZB/4/6<br />

57<br />

Irvine D, Holyoake T,<br />

Copland M.<br />

Holyoake T, Pellicano<br />

F.<br />

Armstrong-Fischer SS,<br />

Urbaniak SJ.<br />

CSO (submitted)<br />

Dr Juraj Petrik (coapplicant):<br />

Prof B Dhillon, Dr K<br />

Ramaesh, Prof M<br />

Turner<br />

Do self renewal pathways<br />

represent a potential therapeutic<br />

target in the treatment of Chronic<br />

Myeloid Leukaemia. Chief<br />

Scientist Office<br />

An investigation of the role of<br />

FOXO transcription factors in CML<br />

stem cell quiescence and as<br />

potential targets in CML stem cell<br />

eradication<br />

Non-Invasive Prenatal Fetal D<br />

typing (follow up on optimisation<br />

assay timing)<br />

Epidemiology and identification of<br />

potential transmission routes of<br />

autochthonous Hepatitis E virus<br />

(HEV) in Scotland and clinical<br />

relevance of HEV in chronic<br />

infection<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£189,257. Jan 2009 – Dec<br />

2011<br />

£223,375. April 2009 –<br />

march 2012<br />

£62, 259 over 12 months<br />

from June 2009<br />

Follow on bid submitted<br />

for ~ £170,000<br />

2-year grant: appr.<br />

£310,000.<br />

<strong>SNBTS</strong> (18 months):<br />

£60,000<br />

76,955 for 2 years<br />

contract under negotiation<br />

Foundation BioAlliance BioSecure<br />

Coste J, Jones M,<br />

Prowse C, Segarra C,<br />

head M, Laude H,<br />

Eglin R, Haik S, Flan<br />

B,<br />

Perret-Liaudet A,<br />

Clewley J.<br />

UK <strong>Blood</strong> Services Forum<br />

Prowse C, Turner M,<br />

Jones M, Head M:<br />

To develop “Screening and<br />

confirmatory testing for<br />

vCJD” on blood samples<br />

vCJD confirmatory assay<br />

developments<br />

<strong>SNBTS</strong> component of Prion filter<br />

trial (PRISM)<br />

<strong>National</strong> Services Scotland (<strong>Scottish</strong> <strong>National</strong> <strong>Blood</strong> <strong>Transfusion</strong> Service)<br />

Prowse C, Turner M,<br />

Jones M, Head M<br />

For infrastructure and routine<br />

support of vCJD assay<br />

development<br />

euro 252,000 over two years<br />

from October 2009 to<br />

September 2011<br />

(126,000 euro pa)<br />

£179,563 over 3 years from<br />

1 st April 2008-2011<br />

(£60,000pa)<br />

2 part time nurses and<br />

patient costs<br />

CVP £65,000 capital and<br />

£80,648 pa salary and<br />

consumables over 3 years<br />

until 31st March 2011.<br />

(£26,882 pa)<br />

SJ Urbaniak, SS<br />

Armstrong-Fisher.<br />

Service Redesign Committee– pilot<br />

study of non-invasive RhD<br />

genotyping on maternal plasma.<br />

£56,480. (2006-2009)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 57 of 74


<strong>Scottish</strong> Enterprise Proof of Concept<br />

Moss M, Urbaniak SJ. <strong>Blood</strong> group mimotopes 248,945<br />

(3 years 2008 to 2010)<br />

Moss M, Urbaniak SJ. <strong>Blood</strong> group mimotopes £90,015.<br />

(1 year 2009-2010)<br />

ITI Life Sciences<br />

Mountford J, Houslay<br />

MD, Milligan G, Baker<br />

AH.<br />

Stem Cells for Safer Medicine<br />

Mountford J, Baker<br />

AH.<br />

Stem Cell Technologies<br />

Programme renewal<br />

Optimisation and functional<br />

validation of directed differentiation<br />

of human embryonic stem cells to<br />

cardiomyocytes.<br />

£762,320 January 2009 –<br />

December 2009.<br />

£66,160. Aug 2009 – July<br />

2010.<br />

Nestech Partnership<br />

SJ Urbaniak, L Cairns,<br />

RN Barker, E Rattray.<br />

Peptide immunotherapy to<br />

suppress antibodies to red blood<br />

cells.<br />

£203,875. (2005-2010)<br />

Polish State Committee for Scientific <strong>Research</strong><br />

Cedzynski M, Atkinson<br />

APM, Swierzko AS,<br />

Szemraj J, MacDonald<br />

SL, Kilpatrick DC,<br />

Buczylko K, Zeman K.<br />

Investigation of the role of<br />

insufficiency of L-Ficolin and other<br />

components of the lectin pathway<br />

of complement activation in<br />

recurrent respiratory infections in<br />

children with allergies<br />

£26,000<br />

(185,000 zl)<br />

(3 years April 2006 to April<br />

2009)<br />

Kay Kendall Leukaemia Fund<br />

Holyoake T, Salmoni<br />

P.<br />

Saudi Arabian Government<br />

Moss M, Urbaniak S<br />

(Abdulla Meshi)<br />

DiaMed AG (Division of Bio-Rad)<br />

R H Fraser<br />

UCB Celltech<br />

S Armstrong-Fisher,<br />

SJ Urbaniak<br />

Alba BioScience<br />

Petrik, Juraj<br />

RH Fraser, CV<br />

Prowse<br />

Targeting autophagy as a tool to<br />

potentiate Imatinib-induced cell<br />

death in CML cells..<br />

Characterisation of the human<br />

humoral immune response to RhD<br />

derived synthetic peptides.<br />

Development of novel monoclonal<br />

blood grouping reagents.<br />

Evaluation of UCB Molecule<br />

Certolizumab Pegol in the human<br />

placental transport in vitro model.<br />

Microarray diagnostics for blood<br />

borne viruses<br />

Development of Anti-Kell Murine<br />

Monoclonal Antibody<br />

£330,216. Proposed start<br />

date 1st October 2009 for 3<br />

years.<br />

£89,745<br />

2008-2011<br />

50,000 SFr per annum<br />

(£22,000) <strong>SNBTS</strong><br />

contribution for 2009 - £4000<br />

Goods & Services (2004-<br />

2010)<br />

2009: £91,861. <strong>SNBTS</strong><br />

contribution for 2009 – Nil<br />

[NB: <strong>SNBTS</strong> share of grant<br />

£28k]<br />

Follow on contract work<br />

agreed<br />

2009: £ ~ 56,000. 2010: £<br />

~ 58,000<br />

2009: £20,628.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 58 of 74


Chemgenex<br />

Holyoake T.<br />

The role of omacetaxine in<br />

targeting cancer stem cells in CML.<br />

Edinburgh University (IKTF fund and CJD surveillance Unit)<br />

Jones M, Prowse C,<br />

Head M.<br />

Salary for technician to<br />

characterise anti-prion monoclonal<br />

antibodies :<br />

£12,000. June 2009 to May<br />

2010;<br />

£18,486 over 12 months to<br />

December 2010<br />

GUHT Endowments<br />

SJ Urbaniak. <strong>Research</strong> Technician £36,000. (2008-2010)<br />

Glasgow Royal Infirmary Endowments<br />

Holyoake TL and Allan<br />

E.<br />

Targeting quiescent CML stem cell<br />

by combining G-CSF and imatinib<br />

mesylate.<br />

TMERF (<strong>Transfusion</strong> Medicine Education and <strong>Research</strong> Fund)<br />

Douglas Watson/ Tim Support for statistical analysis on<br />

Walsh<br />

ISOC (plasma audit)<br />

9,700<br />

(6 years to January 2011)<br />

(2008/ 09) - £15,000<br />

2010-2011<br />

Andrews PD, Becroft M, Aspegren A, Gilmour J, James MJ, McRae S, Kime RA, Allcock RW, Abraham A,<br />

Jiang Z, Strehl R, Mountford JC, Milligan G, Houslay MD, Adams DR, Frearson JA. High content screening<br />

of feeder-free human embryonic stem cells to identify pro-survival small molecules. Biochem J. 2010; 432:<br />

21-33.<br />

Barr PJ, Donnelly M, Morris K, Parker M, Cardwell C, Baillie KEM. The epidemiology of red cell transfusion.<br />

Vox Sanguinis (2010) 99: 239-250<br />

Burton P, Adams DR, Abraham A, Allcock RW, Jiang Z, McCahill A, Gilmour J, McAbney J, Kane NM, Baillie<br />

GS, McKenzie FR, Baker AH, Houslay MD, Mountford JC, Milligan G. Identification and characterization of<br />

small-molecule ligands that maintain pluripotency of human embryonic stem cells. Biochem Soc Trans. 2010<br />

Aug; 38: (4):1058-61<br />

Burton P, A Adams DR, Abraham A, Allcock RW, Jiang Z, McCahill A, Gilmour J, McAbney J, Kaupisch A,<br />

Kane NM, Baillie GS, McKenzie FR, Baker AH, Milligan G, Houslay MD, Mountford JC. Erythro-9-(2-<br />

hydroxy-3-nonyl) Adenine (EHNA) blocks differentiation and maintains the expression of pluripotency<br />

markers in human embryonic stem cells. Biochem. J. Oct 2010<br />

Chalmers JD, Fleming GB, Hill AT, Kilpatrick DC. Impact of mannose-binding lectin insufficiency on the<br />

course of cystic fibrosis: A review and meta-analysis. Glycobiology 2011 Mar; 21: (3) 271-282.<br />

Clark P, Walker ID, Langhorne P, Crichton L, Thomson A, Greaves M, Whyte S, Greer IA. SPIN (<strong>Scottish</strong><br />

Pregnancy Intervention) study: a multicenter, randomised controlled trial of low-molecular-weight heparin and<br />

low-dose aspirin in women with recurrent miscarriage. <strong>Blood</strong>. 2010; 115: (21): 4162-4167.<br />

Dehghan A, Yang Q, Peters A, Basu S, Bis JC, Rudnicka AR, Kavousi M, Chen MH, Baumert J, Lowe GD,<br />

McKnight B, Tang W, de Maat M, Larson MG, Eyhermendy S, McArdle WL, Lumley T, Pankow JS, Hofman<br />

A, Massaro JM, Rivadeneira F, Kolz M, Taylor KD, van Duijn CM, Kathiresan S, Illig T, Aulchenko YS, Volcik<br />

KA, Johnson AD, Uitterlinden AG, Tofler GH, Gieger C; Wellcome Trust Case Control Consortium, Psaty<br />

BM, Couper DJ, Boerwinkle E, Koenig W, O'Donnell CJ, Witteman JC, Strachan DP, Smith NL, Folsom AR.<br />

Wellcome Trust Case Control Consortium. Association of novel genetic Loci with circulating fibrinogen<br />

levels: a genome-wide association study in 6 population-based cohorts. Circ Cardiovasc Genet. 2009; 2:<br />

125-33.<br />

Devine DV, Reesink HW, Panzer S, Irving DO, Kormoczi GF, Mayr WR, Blais Y, Zhu Y, Qian Kk, Zhu Z,<br />

Greinacher A, Grazzini G, Pupella S, Catalano L, Vaglio S, Liumbruno GM, Smeenk JW, Josemans EAJ,<br />

Briet E, Letowska M, Lachert E, Antoniewicz-Papis J, Brojer E, Gulliksson H, Scott M, Williamson L, Prowse<br />

C, AuBuchon JP, Lopez JA, Hoffman P, Busch MP, Norris PJ, Tomasulo P, Dodd RY. <strong>Research</strong> and<br />

development. Vox Sanguinis 2010; 99: 382-401<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 59 of 74


Dijkstra-Tiekstra MJ, van der Meer PF, Cardigan R, Devine D, Prowse C, Sandgren P, and de Wildt-Eggen<br />

J. Platelet concentrates from fresh or overnight-stored blood, an international study. <strong>Transfusion</strong> 2011;<br />

51: 38S-44S.<br />

Estes JD, Harris, LD, Klatt, NR, Tabb B, Pittaluga S, Paiaardini M, Barclay GR, Smedley J, Pung, R, Oliveira<br />

KM, Hirsch VM, Silvestri G, Douek DC, Miller CJ, Haase AT, Lifson J, and Brenchley JM. Damaged<br />

Intestinal Epithelial Integrity Linked to Microbial Translocation in Pathogenic Simian Immunodeficiency Virus<br />

Infections. PLoS Pathogens Aug. 2010 6: Issue 8 e1001052<br />

Fotaki V, Larralde O, Zeng S, McLoughlin D, Nichols J, Price DJ, Theil T, Manson JO. Loss of Wnt8b has no<br />

overt effect on hippocampus development but heads to altered Wnt gene expression levels in dorsomedial<br />

telencephalon. Dev Dyn 2010; 239: 284-296.<br />

Graham D. All water in this establishment has been passed by the management. Medical Device<br />

Decontamination. 2011; 15 (2) 14-18<br />

Grozeva D, Kirov G, Ivanov D, Jones IR, Jones L, Green EK, St Clair DM, Young AH, Ferrier N, Farmer AE,<br />

McGuffin P, Holmans PA, Owen MJ, O'Donovan MC, Craddock N; Wellcome Trust Case Control<br />

Consortium. Rare copy number variants: a point of rarity in genetic risk for bipolar disorder and<br />

schizophrenia. Arch Gen Psychiatry. 2010; 67: 318-27.<br />

Hay, D, Ross J, Gallagher R, Bagnaninchi P. Special Edition: The Complexities of Engineering Human<br />

Stem Cell-Derived Therapeutics. J. Biomed. & Biotech. Volume 2010<br />

Hornsey V, Casey C, McColl K, Young H, Drummond O, McMillan L, Morrison A, Prowse C.<br />

Characteristics of prion filtered red cells suspended in pathogen inactivated plasma (MB treated or solventdetergent<br />

treated) for neonatal exchange transfusion. Vox Sanguinis 2011; 101: 28-34<br />

Jarvis, L.M., Mulligan, K.E., Dunsford, T.H., McGowan, K., Petrik, J. Suitability of an automated nucleic acid<br />

extractor (easyMAG) for use with HCV and HIV-1 NAT assays. J. Virol. Methods 2010. In press. Available<br />

online 30/11/2010.<br />

Jones M, Peden AH, Head MMW, Ironside JW. The application of in vitro cell-free conversion systems to<br />

human prion diseases. Acta Neuropathol. 2010<br />

Kane NM, Meloni M, Spencer HL, Craig MA, Strehl R, Milligan G, Houslay MD, Mountford JC, Emanueli C,<br />

Baker AH. Derivation of endothelial cells from human embryonic stem cells by directed differentiation:<br />

analysis of microRNA and angiogenesis in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2010; Jul; 30:<br />

(7):1389-97. Epub 2010 Apr 29<br />

Khanim FL, Hayden RE, Birtwistle J, Lodi A, Tiziani S, Davies NJ, Ride JP, Viant MR, Gunther UL,<br />

Mountford JC, Schrewe H, Green RM, Murray JA, Drayson MT, Bunce CM. Combined bezafibrate and<br />

medroxyprogesterone acetate: potential novel therapy for acute myeloid leukaemia. PLoS One. 2009 Dec<br />

7;4: (12):e8147<br />

Larralde-Diaz OG, Smith RWP, Malik P, Graham SV, Gray NK. Regulation of poly(A) binding protein during<br />

herpes simplex virus infection. Int. J. Infection. Dis 2010; 14: e470.<br />

Lozano M, Knutson M, Tardivel R, Cid J, Maymo R, Hof HM, Roddie HP, Pelly JP, Docherty AR, Sherman<br />

CD, Lin M, Propst M, Corash L, Prowse CV. A Multi-Centre Study of the Efficacy and Safety of Platelet<br />

Components prepared with Pathogen Inactivation (Intercept TM) stored for 6 or 7 days prior to <strong>Transfusion</strong><br />

(TESSI) Brit. J. Haematol. 2011; 153: 393-401<br />

MacDonald SL, Downing I, Atkinson APM, Gallagher RCJ, Turner ML, Kilpatrick DC. Dendritic cells<br />

previously exposed to mannan-binding lectin (MBL) enhance cytokine production in allogeneic mononuclear<br />

cell cultures. Human Immunology 2010; 71: 1077-1083<br />

Morrison A, McMillan L, Hornsey VS, Prowse CV. Stored red-blood-cells inhibit platelet function under<br />

physiologic flow. Vox Sanguin. 2010 99: 362-368<br />

Mountford JC, Olivier E, Jordanides NE, de Sousa P, Turner ML. Red blood cells from pluripotent stem cells<br />

for use in transfusion. Regen Med. 2010 May; 5: (3): 411-423.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 60 of 74


Mountford J, Olivier E, Turner M. Prospects for the manufacture of red cells for transfusion. Br. J.<br />

Haematology, 2010; 149: (1) 22-34.<br />

Pellicano F, Copland M, Jorgensen HG, Mountford J, Leber B, Holyoake TL. BMS-214662 induces<br />

mitochondrial apoptosis in chronic myeloid leukemia (CML) stem/progenitor cells, including CD34+38- cells,<br />

through activation of protein kinase Cbeta. <strong>Blood</strong>. 2009 Nov 5:114(19):4186-96.<br />

Prowse CV, Murphy WG. Kills 99% of known germs. <strong>Transfusion</strong>. 2010; 50: 1636-1639.<br />

Prowse CV, Cuthbertson B, Turner M. Risk reduction strategies for variant Creutzfeldt-Jakob disease<br />

transmission by UK plasma products. Letters to the Editors - Haemophilia 2011; 17: p.703-720<br />

Reesink HW, Paner S, McQuilten K, Wood EM, Marks DC, Sendel S, Trigo F, Biagini S, Olyntho S, Devine<br />

DV, Mumford I, Cazenave JP, Rasongles P, Garraud O, Richard P, Schooneman F, Vezon G, Radwan RAl,<br />

Brand A, Hervig T, Castro E, Lozano M, Navarro L, Puig L, Almazan C, MacLennan S, Cardigan R, Franklin<br />

IM, Prowse CV. Pathogen inactivation of platelet concentrates. Vox Sanguinis 2010; 99: 85-95.<br />

Reikvam H, Prowse C, Roddie H, Heddle NM, Hervig T. A pilot study of the possibility and the feasibility of<br />

haemoglobin dosing with red blood cells transfusion. Vox Sanguinis. 2010; 99: 71-76.<br />

Reikvam H., van de Watering L., Prowse C., Devine D., Heddle NM., Hervig T. Evaluation of non-invasive<br />

methods for the estimation of haemoglobin content in red blood cell concentrates. Transf. Med. 2011; 21:<br />

145-149<br />

Salaun C, MacDonald AI, Larralde O, Howard L, Lochtie K, Burgess HM, Brook M, Malik P, Gray NK,<br />

Graham SV. Poly (A) Binding Protein 1 (PABP1) relocalisation to the nucleus during HSV-1 infection is<br />

ICP27-independent and does not inhibit virus replication. J. Virology 2010; 84: (17) 8539-8548<br />

Sarab GA, Moss M, Barker RN, Urbaniak SJ. Naturally processed peptides spanning the HPA-1a<br />

polymorphism are efficiently generated and displayed from platelet glycoprotein by HLA-DRB3*0101-positive<br />

antigen-presenting cells. <strong>Blood</strong>. 2009; 114: 1954-1957.<br />

Thom K, Cleland A, Salaskova M, Candotti D, Petrik J. Prevalence and genetic heterogeneity on SEN virus<br />

genotypes D and H in blood donors from Central and Western Europe and West Africa. <strong>Transfusion</strong><br />

Medicine. 2010; 1365-3148.<br />

Tura O, Crawford J, Barclay GR, Samuel K, Hadoke PWF, Roddie H, Davies J, Turner ML. Granulocyte<br />

colony-stimulating factor (G-CSF) depresses angiogenesis in vivo and in vitro: implications for sourcing cells<br />

for vascular regeneration therapy. J. Thrombosis & Haemostasis. 2010; 8: 1614-1623.<br />

Walsh, T.S., Stanworth S., Prescott, RJ., Lee RJ., Watson DM., Wyncoll D. Prevalence, management, and<br />

outcomes of critically ill patients with prothrombin time prolongation in United Kingdom Intensive care units.<br />

Critical Care Med. 2010; 38: (10) 1939-1946<br />

Watson DM., Stanworth SJ., Wyncoll D., McAuley D., Perkins GD., Young D., Biggin KJ., Walsh TS. A<br />

national clinical scenario-based survey of clinicians’ attitudes towards fresh frozen plasma transfusion for<br />

critically ill patients. Transf. Med. 2011; 21: 124-129.<br />

AuBuchon JP. And Prowse CV. Pathogen Inactivation: The Penultimate Paradigm Shift. AABB Press, 2010<br />

ISBN: 978-1-56395-309-5<br />

Prowse CV. United Kingdom: Imported US-Source Methylene-Blue- Treated Plasma (Chapter 10 in<br />

AuBuchon JP, Prowse CV. Pathogen Inactivation: The Penultimate Paradigm Shift. 2010; AABB publications<br />

Tomaskova, J., Labudova, M., Kopacek, J., Patorek, J., Petrik, J., Pastorekova, S. Lymphocytic<br />

choriomeningitis virus (Chapter 67). In “Molecular Detection of Human Viral Pathogens”. (D. Liu ed.), CRC<br />

Press, Taylor & Francis group (Boca Raton, London New York), 2011 735-747.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 61 of 74


<strong>SNBTS</strong> Active Grants 2010-11<br />

Wellcome Trust<br />

Turner ML, Crawford<br />

D, Haque T, Vickers<br />

MA, Forsythe J.<br />

Turner ML, Mountford<br />

J, Forrester L, De<br />

Sousa P, Anstee D,<br />

Courtney A, Murphy<br />

W.<br />

Establishment of a bank of EBV<br />

specific cytotoxic T cells for clinical<br />

treatment of post-transplant<br />

lympho-proliferative disease.<br />

Proof of principle: human<br />

embryonic stem cell derived red<br />

cell concentrates for clinical<br />

transfusion.<br />

[‘<strong>Blood</strong> Pharma’]<br />

£ 431,069<br />

(. Jan 2010 – June 2012)<br />

£2,745,549<br />

(3 years: July 2009 – June 2012)<br />

<strong>Scottish</strong> Funding Council<br />

Wilmut I, Turner M,<br />

Illius A, Savill J<br />

Strategic Development Grant for<br />

Centre of Regenerative Medicine<br />

2,030,000<br />

(4 years August 2006 to July<br />

2010)<br />

<strong>Scottish</strong> Funding Council Horizon Fund<br />

Cell Therapy Group<br />

Glasgow Univ, Heriot<br />

Watt Univ., Edinburgh<br />

Univ. Dundee Univ,<br />

<strong>SNBTS</strong>, Roslin Cells,<br />

SE.<br />

Industrial generation of RCs for<br />

transfusion “New <strong>Blood</strong> Project”<br />

£3.25 million.<br />

Aug. 2011 - 2016<br />

UK Stem Cell Foundation/ Medical <strong>Research</strong> Council/<strong>Scottish</strong> Enterprise/Chief Scientist’s Office<br />

Noble B, Turner ML. Autologous stem cell based<br />

therapies in musculoskeletal<br />

regenerative medicine.<br />

£1,400,000<br />

(2 years May 2008 to May 2010)<br />

CZB/4/6<br />

57<br />

Dhillon B, Ramaesh K,<br />

Turner ML.<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£116,064 over 2 years from 1 st<br />

April 2010 to31st March 2012.<br />

Project runs from Dec 2009 to<br />

31 st Jan 2014<br />

Sir Jules Thorn Charitable Trust<br />

Forbes SJ, Iredale J,<br />

Hayes P, Kluth D,<br />

Turner ML, Bellamy C,<br />

Marshall I.<br />

Stem cell therapy for liver cirrhosis £999,565<br />

(5 years October 2007 to<br />

September 2012)<br />

Medical <strong>Research</strong> Council<br />

Holyoake TL, Brunton<br />

V. Koschmieder S.<br />

Holyoake T,<br />

Koschmieder S,<br />

Calabretta B, Salmoni<br />

P.<br />

Is Bcr-Abl expression relevant for<br />

the survival of cancer stem cells in<br />

chronic myeloid leukaemia?<br />

Is the introduction of autophagy by<br />

tyrosine kinase inhibitors a key<br />

survival mechanism for chromic<br />

myeloid leukaemia stem cells?<br />

£680,000<br />

(3 years June 2007 to May 2010)<br />

£1,117,312. Proposed start date<br />

1st January 2010 for 3 years.<br />

European Union – Public Health Executive Agency<br />

Franklin IM, Hart M, EU Optimal <strong>Blood</strong> Use Project<br />

Arthur E, McClelland<br />

DBL, Pirie E<br />

€890,285 Euros<br />

€500,000 (56.16% EC funded)<br />

(3 years 2007 – 2010)<br />

Leukaemia <strong>Research</strong> Fund<br />

LRF Holyoake TL. Brunton Is Bcr-Abl expression relevant for £28,221 (3 years Oct 2007 - Sept<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 62 of 74


Grant No<br />

06075<br />

V, Koschmider S the survival of cancer stem cells in<br />

chronic myeloid leukaemia?<br />

2010.)<br />

LRF<br />

Grant No<br />

08018<br />

Holyoake TL,<br />

Bartholomew C,<br />

Strathdee G, Melo J.<br />

EVII isoform expression and<br />

knockdown in CML.<br />

£45,000<br />

(3 years April 2008 to March<br />

2011)<br />

LRF<br />

Grant No<br />

08071<br />

Whetton T,<br />

Holyoake T. A<br />

Cancer <strong>Research</strong> United Kingdom.<br />

Holyoake T, Whetton<br />

T, Girolami M.<br />

phosphoproteomic investigation of<br />

key BCR-ABL mediated signally<br />

pathways in CML stem cells: where<br />

does tyrosine kinase inhibitor<br />

resistance lie?.<br />

Key survival pathways in chronic<br />

myeloid leukaemia (CML) stem<br />

cells and novel approaches to their<br />

eradication. Cancer <strong>Research</strong><br />

United Kingdom.<br />

Biotechnology, Biophysical Scientific <strong>Research</strong> Council (BBSRC)<br />

De Sousa P, Turner<br />

ML, Pells S.<br />

A novel characterisation and<br />

separation technique for pluripotent<br />

human embryonic and<br />

haematopoietic stem cells.<br />

£353,479 Jan 09 – Dec 2011<br />

£1.1 million. Proposed start date<br />

1st January 2010 for 5 years.<br />

Amount applied for £1,138,122.<br />

£131,765<br />

(3 years January 2009 to<br />

December 2012)<br />

British Heart Foundation<br />

BHF<br />

Project<br />

Grant No<br />

PG/06/0<br />

51<br />

BHF<br />

Project<br />

Grant No<br />

PG/07/0<br />

17/2240<br />

5<br />

Barclay GR, Mills N,<br />

Newby D, Hadoke P,<br />

Turner ML.<br />

Mills NL, Newby DE,<br />

Barclay GR, de Belder<br />

MA, Robinson SD<br />

Preclinical in vivo evaluation of<br />

potential sources of human<br />

endothelial progenitor cells for<br />

autograft cellular therapy of<br />

ischaemia<br />

Endothelial progenitor cells in<br />

acute vascular injury and repair.<br />

£239,930<br />

(3 years July 2006 to June 2009)<br />

- extended to end June 2010<br />

(i.e. 4 years total) because of 2<br />

sets of maternity leave by O<br />

Tura).<br />

£242,685<br />

(3 years August 2007 to July<br />

2010)<br />

Department of Health<br />

Manson J, Houston F,<br />

Prowse C, Turner M,<br />

MacGregor I, Hornsey<br />

V, Hunter N,<br />

Foster J, Groschup M.<br />

The effect of leucodepletion on<br />

transmission of BSE by transfusion<br />

of sheep blood components.<br />

Chief Scientist Office – <strong>Scottish</strong> Executive Health Department (CSO SEHD)<br />

Head MW, Peden A,<br />

Prowse C, Manson J,<br />

Bishop M, Ironside J.<br />

Grant No<br />

CAF/08/<br />

09.<br />

Walsh T, Forbes J,<br />

Langston A,<br />

McClelland DBL, Pelly<br />

J, Ramsay P, Watson<br />

D, Prowse C.<br />

Irvine D, Holyoake T,<br />

Copland M.<br />

The use of human recombinant<br />

prion protein as a substrate in an<br />

improved in vitro amplificationbased<br />

blood test for Creutzfeldt-<br />

Jakob disease infectivity<br />

A feasibility randomized trial<br />

comparing REstrictive and LIberal<br />

blood transfusion strategies in<br />

patients requiring six or more days<br />

in intEnsiVE care (RELIEVE study)<br />

Do self renewal pathways<br />

represent a potential therapeutic<br />

target in the treatment of Chronic<br />

Myeloid Leukaemia. Chief<br />

Scientist Office<br />

£3,424,382 (6 years to<br />

December 2010)<br />

Extension to 2012 agreed with<br />

DoH & Jean Manson<br />

£193,512 over 2 years to<br />

December 2010 (£90,000pa)<br />

£182,577 over 18 months to<br />

October 2010<br />

(£100,000pa)<br />

£189,257. Jan 2009 – Dec 2011<br />

Grant no Holyoake T, Pellicano An investigation of the role of £223,375. April 2009 – March<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 63 of 74


CZB/4/6<br />

90.<br />

F. FOXO transcription factors in CML<br />

stem cell quiescence and as<br />

potential targets in CML stem cell<br />

eradication<br />

Armstrong-Fischer SS,<br />

Urbaniak SJ.<br />

CSO (submitted)<br />

Dr Juraj Petrik (coapplicant):<br />

Non-Invasive Prenatal Fetal D<br />

typing (follow up on optimisation<br />

assay timing)<br />

Epidemiology and identification of<br />

potential transmission routes of<br />

autochthonous Hepatitis E virus<br />

(HEV) in Scotland and clinical<br />

relevance of HEV in chronic<br />

infection<br />

2012<br />

£62, 259 over 12 months from<br />

June 2009<br />

Follow on bid submitted for ~<br />

£170,000<br />

2-year grant: appr. £310,000.<br />

<strong>SNBTS</strong> (18 months): £60,000<br />

CZB/4/6<br />

57<br />

Prof B Dhillon, Dr K<br />

Ramaesh, Prof M<br />

Turner<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£116,064 over 2 years from 1 st<br />

April 2010 to31st March 2012.<br />

Project runs from Dec 2009 to<br />

31 st Jan 2014<br />

Foundation BioAlliance BioSecure<br />

Coste J, Jones M,<br />

Prowse C, Segarra C,<br />

head M, Laude H,<br />

Eglin R, Haik S, Flan<br />

B,<br />

Perret-Liaudet A,<br />

Clewley J.<br />

To develop “Screening and<br />

confirmatory testing for<br />

vCJD” on blood samples<br />

euro 252,000 over two years<br />

from October 2009 to September<br />

2011<br />

(126,000 euro pa)<br />

UK <strong>Blood</strong> Services Forum<br />

Prowse C, Turner M,<br />

Jones M, Head M:<br />

vCJD confirmatory assay<br />

developments<br />

<strong>SNBTS</strong> component of Prion filter<br />

trial (PRISM)<br />

£179,563 over 3 years from 1 st<br />

April 2008-2011 (£60,000pa)<br />

2 part time nurses and patient<br />

costs<br />

<strong>National</strong> Services Scotland (<strong>Scottish</strong> <strong>National</strong> <strong>Blood</strong> <strong>Transfusion</strong> Service)<br />

Prowse C, Turner M,<br />

Jones M, Head M<br />

For infrastructure and routine<br />

support of vCJD assay<br />

development<br />

CVP £65,000 capital and<br />

£80,648 pa salary and<br />

consumables over 3 years until<br />

31st March 2011. (£26,882 pa)<br />

<strong>Scottish</strong> Enterprise Proof of Concept<br />

Moss M, Urbaniak SJ. <strong>Blood</strong> group mimotopes 248,945<br />

(3 years 2008 to 2010)<br />

Moss M, Urbaniak SJ. <strong>Blood</strong> group mimotopes £90,015.<br />

(1 year 2009-2010)<br />

Stem Cells for Safer Medicine.<br />

Mountford J, Baker<br />

AH.<br />

Nestech Partnership<br />

SJ Urbaniak, L Cairns,<br />

RN Barker, E Rattray.<br />

Optimisation and functional<br />

validation of directed differentiation<br />

of human embryonic stem cells to<br />

cardiomyocytes.<br />

Peptide immunotherapy to<br />

suppress antibodies to red blood<br />

cells.<br />

£66,160. Aug 2009 – July 2010.<br />

£203,875. (2005-2010)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 64 of 74


Kay Kendall Leukaemia Fund<br />

Holyoake T, Salmoni<br />

P.<br />

Saudi Arabian Government<br />

Moss M, Urbaniak S<br />

(Abdulla Meshi)<br />

DiaMed AG (Division of Bio-Rad)<br />

R H Fraser<br />

UCB Celltech<br />

S Armstrong-Fisher,<br />

SJ Urbaniak<br />

Alba BioScience<br />

Petrik, Juraj<br />

Chemgenex<br />

Holyoake T.<br />

Targeting autophagy as a tool to<br />

potentiate Imatinib-induced cell<br />

death in CML cells.<br />

Characterisation of the human<br />

humoral immune response to RhD<br />

derived synthetic peptides.<br />

Development of novel monoclonal<br />

blood grouping reagents.<br />

Evaluation of UCB Molecule<br />

Certolizumab Pegol in the human<br />

placental transport in vitro model.<br />

Microarray diagnostics for blood<br />

borne viruses<br />

The role of omacetaxine in<br />

targeting cancer stem cells in CML.<br />

£330,216. Proposed start date<br />

1st October 2009 for 3 years.<br />

£89,745<br />

2008-2011<br />

50,000 SFr per annum (£22,000)<br />

<strong>SNBTS</strong> contribution for 2009 -<br />

£4000 Goods & Services (2004-<br />

2010)<br />

2009: £91,861. <strong>SNBTS</strong><br />

contribution for 2009 – Nil [NB:<br />

<strong>SNBTS</strong> share of grant £28k]<br />

Follow on contract work<br />

agreed<br />

2009: £ ~ 56,000. 2010: £ ~<br />

58,000<br />

£12,000. June 2009 to May<br />

2010;<br />

Edinburgh University (IKTF fund and CJD surveillance Unit)<br />

Jones M, Prowse C,<br />

Head M.<br />

Salary for technician to<br />

characterise anti-prion monoclonal<br />

antibodies :<br />

£18,486 over 12 months to<br />

December 2010<br />

GUHT Endowments<br />

SJ Urbaniak. <strong>Research</strong> Technician £36,000. (2008-2010)<br />

Glasgow Royal Infirmary Endowments<br />

Holyoake TL and Allan<br />

E.<br />

Targeting quiescent CML stem cell<br />

by combining G-CSF and imatinib<br />

mesylate.<br />

£9,700 (6 years to January 2011)<br />

2011-2012<br />

Allan EK, Holyoake TL, Craig AR, Jorgensen HB. Omacetaxine may have a role in chronic myeloid<br />

leukaemia eradication through down-regulation of Mcl-1 and induction of apoptosis in stem/progenitor cells.<br />

Leukemia 2011 Apr 5 In press.<br />

Balabanov S, Gontarewicz A, Keller G, Raddrizzani L, Braig M, Bosotti R, Moll J, Jost E, Barett C, Rohe I,<br />

Bokemeyer C, Holyoake TL, Brümmendorf TH. Abcg2 overexpression represents a novel mechanism for<br />

acquired resistance to the multi-kinase inhibitor Danusertib in BCR-ABL-positive cells in vitro. PLoS One.<br />

2011 Apr 26;6(4):e19164. PubMed PMID: 21541334; PubMed Central PMCID: PMC3082549.<br />

Baylis SA, heath AB, and the Collaborative Study Group* (including Jarvis L and Cleland A). World Health<br />

Organisation collaborative study to calibrate the 3 rd International Standard for Hepatitis C virus RNA nucleic<br />

acid amplification technology (NAT)-based assays. Vox sang. 2011; epub ahead of print.<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 65 of 74


Bessos, H. <strong>Transfusion</strong> and Apheresis Science 2011: 45, 29<br />

Bessos, H. Fraser, R. Seghatchian, J. Scotblood 2010: Key presentations of the past, present, and future of<br />

transfusion medicine to mark <strong>Scottish</strong> national blood transfusion service (<strong>SNBTS</strong>) anniversaries.<br />

<strong>Transfusion</strong> and Apheresis Science 2011: 45: 213-221.<br />

Bessos, H., Fraser, R., Seghatchian, J. Scotblood 2011 features advances in translational medicine,<br />

haemophoietic progenitor cells and milestones of blood banking systems. <strong>Transfusion</strong> and Apheresis<br />

Science 2011: 445: 315-320.<br />

Chalmers JD, Fleming GB, Hill AT, Kilpatrick DC. Impact of mannose-binding lectin insufficiency on the<br />

course of cystic fibrosis: A review and meta-analysis. Glycobiology. 2011 Mar;21(3):271-82. Epub 2010 Nov<br />

2. Review. PubMed PMID: 21045008.<br />

Crowther M, Chan YLT, Garbett IK, Lim W, Vickers MA, and Crowther MA (2011) Evidence based focused<br />

review of the treatment of idiopathic warm IgG hemolytic anemia in adults. <strong>Blood</strong> 118:4036-40.<br />

Farrugia A, Prowse, C, Hornsey, V. Conditions for plasma processing. Letters to the Editor, <strong>Transfusion</strong>.<br />

2011: 51: p.1875-76<br />

Gallipoli P, Abraham SA, Holyoake TL. Hurdles toward a cure for CML: the CML stem cell. Hematol Oncol<br />

Clin North Am. 2011 Oct;25(5):951-66, v. Epub 2011 Oct 19. PubMed PMID: 22054728.<br />

Hall AM, Zamzani OM, Whibley N, Hampsey DP, Haggart AM, Vickers MA, Barker RN (2012) Production of<br />

the effector cytokine interleukin-17, rather than interferon- is more strongly associated with autoimmune<br />

haemolytic anemia. The Haematology Journal (in press)<br />

Hansen A, McMillan L, Morrison A, Petrik J, Bradley M. Polymers for the rapid and effective activation and<br />

aggregation of platelets. Biomaterials 2011; 32:7034-41.<br />

Hardie, I. Rooney, C. The increasing importance of intellectual property in transfusion medicine. <strong>Transfusion</strong><br />

and Apheresis Science 2011: 45: 99-107.<br />

Hay DC, Ross JA, Gallagher R, Bagnaninchi P. The complexities of engineering human stem cell-derived<br />

therapeutics. J Biomed Biotechnol. 2010;2010:654964. Epub 2011 Mar 31. PubMed PMID: 21490704;<br />

PubMed Central PMCID: PMC3070168.<br />

Hornsey VS, Casey C, McColl K, Young H, Drummond O, McMillan L, Morrison A and Prowse CV (2011)<br />

Characteristics of prion-filtered red cells suspended in pathogen-inactivated plasma (MB treated or solventdetergent<br />

treated) for neonatal exchange transfusion. Vox Sang. 101, 28-34<br />

Ibrahim AR, Clark RE, Holyoake TL, Byrne J, Shepherd P, Apperley JF,<br />

Milojkovic D, Szydlo R, Goldman J, Marin D. Second-generation tyrosine kinase inhibitors improve the<br />

survival of patients with chronic myeloid leukemia in whom imatinib therapy has failed. Haematologica. 2011<br />

Dec;96(12):1779-82. Epub 2011 Aug 22. PubMed PMID: 21859733; PubMed Central PMCID: PMC3232259.<br />

Ivanovs, A, Rybtsov, S., Welch, l., Anderson, R.A., Turner, M.L., Medvinsky, A. Highly potent human<br />

hematopoietic stem cells first emerge in the intraembryonic aorta-gonad-mesonephros region. J. Exp.Med.<br />

2011: 208: No. 12; 2417-2427<br />

Jarvis LM, Mulligan K, Dunsford TH, McGowan K, Petrik J. Suitability of an automated nucleic acid extractor<br />

(easyMAG) for use with hepatitis C virus and human immunodeficiency virus type 1 nucleic acid amplification<br />

testing. J Virol Methods 2011; 171:364-8.<br />

Krejciova Z, Pells S, Cancellotti E, Freile P, Bishop M, Samuel K, Barclay RG, Ironside JW, Manson JC,<br />

Turner ML, De Sousa P, Head MW. Human embryonic stem cells rapidly take up and then clear exogenous<br />

human and animal prions in vitro. J. Pathol. 2011 Apr; 223 (5): 635-645.<br />

Larralde-Diaz OG, Smith RWP, Malik P, Graham SV, Gray NK. Regulation of poly(A) binding protein during<br />

herpes simplex virus infection. Int. J. Infection Dis. 2010; 14: e470.<br />

Lidonnici MR, Audia A, Soliera AR, Prisco M, Ferrari-Amorotti G, Waldron T, Donato N, Zhang Y, Martinez<br />

RV, Holyoake TL, Calabretta B. Expression of the transcriptional repressor Gfi-1 is regulated by<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 66 of 74


C/EBP{alpha} and is involved in its proliferation and colony formation-inhibitory effects in p210BCR/ABLexpressing<br />

cells. Cancer Res. 2010 Oct 15;70(20):7949-59. Epub 2010 Oct 5. PubMed PMID: 20924107;<br />

PubMed Central PMCID: PMC2955805.<br />

MacDonald SL, Downing I, Atkinson AP, Gallagher RC, Turner ML, Kilpatrick DC. Dendritic cells previously<br />

exposed to mannan-binding lectin enhance cytokine production in allogeneic mononuclear cell cultures. Hum<br />

Immunol. 2010 Nov;71(11):1077-83. Epub 2010 Aug 9. PubMed PMID: 20705112.<br />

Marenah L, Allan EK, Mountford JC, Holyoake TL, Jørgensen HG, Elliott MA. Investigation into omacetaxine<br />

solution stability for in vitro study. Biomed Chromatogr. 2011 Aug 9. doi: 10.1002/bmc.1686. [Epub ahead of<br />

print] PubMed PMID:21830228.<br />

Martineau HM, Cousens C, Imlach S, Dagleish MP, Griffiths DJ. Jaagsiekte sheep<br />

retrovirus infects multiple cell types in the ovine lung. J Virol. 2011 Apr;85(7):3341-55. Epub 2011 Jan 26.<br />

PubMed PMID: 21270155; PubMed Central PMCID:PMC3067841.<br />

McCutcheon S, Blanco A.R.A, Houston E.F, de Wolf, C, Tan, B.C, Smith, A, Groschup M.H, Hunter, N,<br />

Hornsey, V.S, MacGregor, I.R, Prowse, C.V, Turner, M. Manson, J.C. All clinically-relevant blood<br />

components transmit prion disease following a single blood transfusion: A sheep model of vCJD. PLoS<br />

One, 2011: 6: Issue 8, 1-11 e23169.<br />

Metcalfe P, Rigsby P, Tait E, Urbaniak SJ (2011) An international reference reagent for the detection of RHD<br />

and SRY DNA in plasma. Vox Sang Epub 2011 Oct 4. DOI:10.1111/j.1423-0410.2011.01543.x<br />

Milliken SVI, Wassall H, Lewis BJ, Logie J, Barker RN, Macdonald H, Vickers MA, Ormerod AD (2012)<br />

Effects of ultraviolet light on human serum 25-hydroxyvitamin D and systemic immune function. Journal of<br />

Allergy and Clinical Immunology, 129, 1554-61<br />

Mitchell SA, Jacobsohn D, Thormann Powers KE, Carpenter PA, Flowers ME, Cowen EW, Schubert M,<br />

Turner ML, Lee SJ, Martin P, Bishop MR, Baird K, Bolaños-Meade J, Boyd K, Fall-Dickson JM, Gerber LH,<br />

Guadagnini JP, Imanguli M, Krumlauf MC, Lawley L, Li L, Reeve BB, Clayton JA, Vogelsang GB, Pavletic<br />

SZ. A multicenter pilot evaluation of the <strong>National</strong> Institutes of Health chronic graft-versus-host disease<br />

(cGVHD) therapeutic response measures: feasibility, interraterreliability, and minimum detectable change.<br />

Biol <strong>Blood</strong> Marrow Transplant. 2011 Nov;17(11):1619-29. Epub 2011 Apr 12. PubMed PMID: 21536143;<br />

PubMed Central PMCID: PMC3158826.<br />

Mountford JC, Turner, M. In vitro production of red blood cells. <strong>Transfusion</strong> and Apheresis Science 2011:<br />

45: 85-89.<br />

Mukhopadhyay A, Helgason GV, Karvela M, Holyoake TL. Hydroxychloroquine for chronic myeloid leukemia:<br />

complete cure on the horizon? Expert Rev Hematol. 2011 Aug;4(4):369-71. PubMed PMID: 21801126.<br />

Helgason GV, Karvela M, Holyoake TL. Kill one bird with two stones: potential efficacy of BCR-ABL and<br />

autophagy inhibition in CML. <strong>Blood</strong>. 2011 Aug 25;118(8):2035-43. Epub 2011 Jun 21. Review. PubMed<br />

PMID: 21693757.<br />

Nightingale MJ, De Korte D, Chabanel, A, Hughes, W, Rowe, GP, Nicholson G. Eurobloodpack: a common<br />

European design for blood bag systems with integral Leucodepletion filters. Vox Sang. Online 18/04/2011<br />

doi: 10.1111/j.1423-0410.2011.01480.<br />

Olivier EN, Bouhassira EE. Differentiation of human embryonic stem cells into mesenchymal stem cells by<br />

the "raclure" method. Methods Mol Biol. 2011;690:183-93. PubMed PMID: 21042994<br />

Payne CM, Samuel K, Pryde A, King J, Brownstein D, Schrader J, Medine CN,Forbes SJ, Iredale JP,<br />

Newsome PN, Hay DC. Persistence of functional hepatocyte-like cells in immune-compromised mice. Liver<br />

Int. 2011 Feb;31(2):254-62. doi: 10.1111/j.1478-3231.2010.02414.x. Epub 2010 Dec 10. PubMed PMID:<br />

21143581.<br />

Pellicano F, Holyoake TL. Assembling defenses against therapy-resistant leukemic stem cells: Bcl6 joins the<br />

ranks. J Exp Med. 2011 Oct 24;208(11):2155-8. PubMed PMID: 22025499; PubMed Central PMCID:<br />

PMC3201197.<br />

Pellicano F, Simara P, Sinclair A, Helgason GV, Copland M, Grant S, Holyoake TL. The MEK inhibitor<br />

PD184352 enhances BMS-214662-induced apoptosis in CD34+ CML stem/progenitor cells. Leukemia. 2011<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 67 of 74


Jul;25(7):1159-67. doi:10.1038/leu.2011.67. Epub 2011 Apr 12. PubMed PMID: 21483442<br />

Pellicano F, Sinclair A, Holyoake TL. In search of CML stem cells' deadly weakness. Curr Hematol Malig<br />

Rep. 2011 Jun;6(2):82-7. Review. PubMed PMID: 21373837.<br />

Petrik J. Microarrays for blood testing: pros and cons. Biologicals 2010; 38: 2-8.<br />

Petrik J, Coste J, Fournier-Wirth C. Advances in transfusion medicine in the first decade of the 21st century:<br />

Advances in miniaturized technologies. Transfus Apher Sci. 2011; 45:45-51.<br />

Prowse C, Cuthbertson, B, Turner, M. Risk reduction strategies for variant Creutzfeldt-Jakob disease<br />

transmission by UK plasma products. Haemophilia 2011: 17: 703-720<br />

Stephen J, Cairns LS, Pickford WJ, Vickers MA, Urbaniak SJ and Barker RN (2012) Identification,<br />

immunomodulatory activity and immunogenicity of the major helper T cell epitope on the K blood group<br />

antigen. <strong>Blood</strong>, 119, 5563-74<br />

Stuge, T.B. Skogen B, Ahlen, M.T., Husebekk, A., Urbaniak, S.J., Bessos, H. The cellular immunobiology<br />

associated with fetal and neonatal alloimmune thrombocytopenia. <strong>Transfusion</strong> and Apheresis Science.<br />

2011: 45: p.53-59.<br />

Thom K, Cleland A, Salakova M, Candotti D, Petrik J: Prevalence and genetic heterogeneity of SEN virus<br />

genotypes D and H in blood donors from Central and Western Europe and West Africa. Transfus Med 2011;<br />

21:42-50.<br />

Tomaskova J, Labudova M, Kopacek J, Pastorek J, Petrik J, Pastorekova S. Lymphocytic choriomeningitis<br />

virus. In: “Molecular detection of human viral pathogens” Chapter 67. CRC Press, Taylor & Francis group<br />

(Boca Ratan, London, new York), 2011 p. 735-747.<br />

Wagner S.J. Pathogen Inactivation: The Penultimate Paradigm Shift. Ed. AuBuchon JP, Prowse CV.<br />

<strong>Transfusion</strong> Medicine Reviews Vol. 25 No: 2 April 2011 p.162<br />

Walsh TS, Palmer J, Watson D, Biggin K, Seretny M, Davidson H, Harkness M, Hay A. Multicentre cohort<br />

study of red blood cell use for revision hip arthroplasty and factors associated with greater risk of allogeneic<br />

blood transfusion. Brit. J. Anaesthesia 2012; 108: (1) 63-71.<br />

Watkins NA, Dobra S, Bennett P, Cairns J, Turner ML. The Management of <strong>Blood</strong> Safety in the Presence of<br />

Uncertain Risk: A United Kingdom Perspective. Transfus Med Rev. 2011 Nov 28. [Epub ahead of print]<br />

PubMed PMID: 22126710.<br />

Yung S, Ledran M, Moreno-Gimeno I, Conesa A, Montaner D, Dopazo J, Dimmick I,Slater NJ, Marenah L,<br />

Real PJ, Paraskevopoulou I, Bisbal V, Burks D, Santibanez-Koref M, Moreno R, Mountford J, Menendez P,<br />

Armstrong L, Lako M. Large-scale transcriptional profiling and functional assays reveal important roles for<br />

Rho-GTPase signalling and SCL during haematopoietic differentiation of human embryonic stem cells. Hum<br />

Mol Genet. 2011 Dec 15;20(24):4932-46. Epub 2011 Sep 21. PubMed PMID: 21937587.<br />

<strong>SNBTS</strong> Active Grants 2011-12<br />

Wellcome Trust<br />

Turner ML, Crawford<br />

D, Haque T, Vickers<br />

MA, Forsythe J.<br />

Turner ML, Mountford<br />

J, Forrester L, De<br />

Sousa P, Anstee D,<br />

Courtney A, Murphy<br />

W.<br />

Establishment of a bank of EBV<br />

specific cytotoxic T cells for clinical<br />

treatment of post-transplant<br />

lympho-proliferative disease.<br />

Proof of principle: human<br />

embryonic stem cell derived red<br />

cell concentrates for clinical<br />

transfusion.<br />

[‘<strong>Blood</strong> Pharma’]<br />

£ 431,069<br />

(Jan 2010 – June 2012)<br />

£2,745,549<br />

(3 years: July 2009 – June 2012)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 68 of 74


Vickers M, Barker R<br />

Erwig L<br />

Immune consequences of red cell<br />

aging, death and disposal<br />

£1,252,158<br />

(5 years Jan 2012-2017)<br />

<strong>Scottish</strong> Funding Council Horizon Fund<br />

Cell Therapy Group<br />

Glasgow Univ, Heriot<br />

Watt Univ., Edinburgh<br />

Univ. Dundee Univ,<br />

<strong>SNBTS</strong>, Roslin Cells,<br />

SE.<br />

Industrial generation of RCs for<br />

transfusion “New <strong>Blood</strong> Project”<br />

£3.25 million.<br />

Aug. 2011 - 2016<br />

UK Stem Cell Foundation/ Medical <strong>Research</strong> Council/<strong>Scottish</strong> Enterprise/Chief Scientist’s Office<br />

CZB/4/6<br />

57<br />

Dhillon B, Ramaesh K,<br />

Turner ML.<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£116,064 over 2 years from 1 st<br />

April 2010 to31st March 2012.<br />

Project runs from Dec 2009 to<br />

31 st Jan 2014<br />

Sir Jules Thorn Charitable Trust<br />

Forbes SJ, Iredale J,<br />

Hayes P, Kluth D,<br />

Turner ML, Bellamy C,<br />

Marshall I.<br />

Stem cell therapy for liver cirrhosis £999,565<br />

(5 years October 2007 to<br />

September 2012)<br />

Medical <strong>Research</strong> Council<br />

Holyoake T,<br />

Koschmieder S,<br />

Calabretta B, Salmoni<br />

P.<br />

Leukaemia <strong>Research</strong> Fund<br />

LRF<br />

Grant No<br />

08071<br />

Whetton T,<br />

Holyoake T. A<br />

Is the introduction of autophagy by<br />

tyrosine kinase inhibitors a key<br />

survival mechanism for chromic<br />

myeloid leukaemia stem cells?<br />

phosphoproteomic investigation of<br />

key BCR-ABL mediated signally<br />

pathways in CML stem cells: where<br />

does tyrosine kinase inhibitor<br />

resistance lie?.<br />

£1,117,312. Proposed start date<br />

1st January 2010 for 3 years.<br />

£353,479 Jan 09 – Dec 2011<br />

Cancer <strong>Research</strong> United Kingdom.<br />

Holyoake T, Whetton<br />

T, Girolami M.<br />

Key survival pathways in chronic<br />

myeloid leukaemia (CML) stem<br />

cells and novel approaches to their<br />

eradication. Cancer <strong>Research</strong><br />

United Kingdom.<br />

Biotechnology, Biophysical Scientific <strong>Research</strong> Council (BBSRC)<br />

De Sousa P, Turner<br />

ML, Pells S.<br />

A novel characterisation and<br />

separation technique for pluripotent<br />

human embryonic and<br />

haematopoietic stem cells.<br />

£1.1 million. Proposed start date<br />

1st January 2010 for 5 years.<br />

Amount applied for £1,138,122.<br />

£131,765<br />

(3 years January 2009 to<br />

December 2012)<br />

Department of Health<br />

Manson J, Houston F,<br />

Prowse C, Turner M,<br />

MacGregor I, Hornsey<br />

V, Hunter N,<br />

Foster J, Groschup M.<br />

The effect of leucodepletion on<br />

transmission of BSE by transfusion<br />

of sheep blood components.<br />

£3,424,382 (6 years to<br />

December 2010)<br />

Extension to 2012 agreed with<br />

DoH & Jean Manson<br />

Chief Scientist Office – <strong>Scottish</strong> Executive Health Department (CSO SEHD)<br />

Grant No<br />

CAF/08/<br />

09.<br />

Irvine D, Holyoake T,<br />

Copland M.<br />

Do self renewal pathways<br />

represent a potential therapeutic<br />

target in the treatment of Chronic<br />

£189,257. Jan 2009 – Dec 2011<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 69 of 74


Grant no<br />

CZB/4/6<br />

90.<br />

Holyoake T, Pellicano<br />

F.<br />

Armstrong-Fischer SS,<br />

Urbaniak SJ.<br />

CSO (submitted)<br />

Scobie L. et al<br />

Dr Juraj Petrik (coapplicant):<br />

Myeloid Leukaemia. Chief<br />

Scientist Office<br />

An investigation of the role of<br />

FOXO transcription factors in CML<br />

stem cell quiescence and as<br />

potential targets in CML stem cell<br />

eradication<br />

Non-Invasive Prenatal Fetal D<br />

typing (follow up on optimisation<br />

assay timing)<br />

Epidemiology and identification of<br />

potential transmission routes of<br />

autochthonous Hepatitis E virus<br />

(HEV) in Scotland and clinical<br />

relevance of HEV in chronic<br />

infection<br />

£223,375. April 2009 – March<br />

2012<br />

£62,259 over 12 months from<br />

June 2009<br />

Follow on bid submitted for ~<br />

£170,000<br />

2-year grant: appr. £310,000.<br />

<strong>SNBTS</strong> (18 months): £60,000<br />

Start October 2010<br />

CZB/4/6<br />

57<br />

Prof B Dhillon, Dr K<br />

Ramaesh, Prof M<br />

Turner<br />

Ex vivo expanded corneal limbal<br />

stem cell transplantation: from<br />

laboratory to clinical application<br />

£116,064 over 2 years from 1 st<br />

April 2010 to31st March 2012.<br />

Project runs from Dec 2009 to<br />

31 st Jan 2014<br />

Foundation BioAlliance BioSecure<br />

Coste J, Jones M,<br />

Prowse C, Segarra C,<br />

head M, Laude H,<br />

Eglin R, Haik S, Flan<br />

B,<br />

Perret-Liaudet A,<br />

Clewley J.<br />

To develop “Screening and<br />

confirmatory testing for<br />

vCJD” on blood samples<br />

Petrik J. SYNPOLYVIR 43,000 Euros<br />

2011-2012<br />

euro 252,000 over two years<br />

from October 2009 to September<br />

2011<br />

(126,000 euro pa)<br />

UK <strong>Blood</strong> Services Forum<br />

Prowse C, Turner M,<br />

Jones M, Head M:<br />

vCJD confirmatory assay<br />

developments<br />

<strong>SNBTS</strong> component of Prion filter<br />

trial (PRISM)<br />

£179,563 over 3 years from 1 st<br />

April 2008-2011 (£60,000pa)<br />

2 part time nurses and patient<br />

costs<br />

<strong>Scottish</strong> Enterprise Proof of Concept<br />

Moss M, Urbaniak SJ. <strong>Blood</strong> group mimotopes £248,945<br />

(3 years 2008 to 2011)<br />

Kay Kendall Leukaemia Fund<br />

Holyoake T, Salmoni<br />

P.<br />

Saudi Arabian Government<br />

Moss M, Urbaniak S<br />

(Abdulla Meshi)<br />

UCB Celltech<br />

S Armstrong-Fisher,<br />

SJ Urbaniak<br />

Targeting autophagy as a tool to<br />

potentiate Imatinib-induced cell<br />

death in CML cells.<br />

Characterisation of the human<br />

humoral immune response to RhD<br />

derived synthetic peptides.<br />

Evaluation of UCB Molecule<br />

Certolizumab Pegol in the human<br />

placental transport in vitro model.<br />

£330,216. Proposed start date<br />

1st October 2009 for 3 years.<br />

£89,745<br />

2008-2011<br />

2009: £91,861. <strong>SNBTS</strong><br />

contribution for 2009 – Nil [NB:<br />

<strong>SNBTS</strong> share of grant £28k]<br />

Follow on contract work agreed<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 70 of 74


Alba BioScience<br />

Petrik, Juraj<br />

Microarray diagnostics for blood<br />

borne viruses<br />

2009-10: £ ~ 56,000.<br />

2010-11: £ ~ 58,000<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 71 of 74


HEALTHCARE QUALITY IMPACT ASSESSMENT FOR SERVICE REDESIGN TEMPLATE Appendix 6<br />

Which health<br />

impacts could<br />

the project<br />

achieve?<br />

(tick all that<br />

apply)<br />

Estimate amount<br />

of impact on<br />

patient/population<br />

small +<br />

medium ++<br />

high +++;<br />

or quantify where<br />

possible<br />

How will the project achieve this<br />

health impacts?<br />

What is the<br />

evidence base for<br />

this?<br />

Is it possible to<br />

define the improved<br />

health outcome for<br />

the patient<br />

Are there key<br />

dependencies in<br />

achieving the<br />

health impact?<br />

What measures<br />

of health<br />

impact could<br />

be used?<br />

INDIRECT<br />

HEALTH<br />

IMPACTS<br />

Safer<br />

More<br />

effective<br />

More<br />

efficient<br />

More<br />

equitable<br />

More<br />

timely<br />

More<br />

person<br />

centred<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 72 of 74


What<br />

health<br />

outcomes<br />

could the<br />

project<br />

achieve?<br />

How will the project achieve these<br />

health outcomes?<br />

What is the<br />

evidence base for<br />

this?<br />

Are there key dependencies in<br />

achieving the health outcome?<br />

What measures of<br />

health outcomes<br />

could be used?<br />

DIRECT<br />

HEALTH<br />

IMPACT<br />

(to be used for<br />

direct patient<br />

care)<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 73 of 74


DESCRIPTIONS OF INDIRECT HEALTH IMPACTS - CONTRIBUTIONS TO THE QUALITY OF CARE<br />

Safe<br />

Effective<br />

Efficient<br />

Equitable<br />

Timely<br />

Person<br />

centred<br />

avoiding injuries to patients from care that is intended to help them; providing an appropriate, clean and safe<br />

environment; reducing harm from infectious and environmental hazards<br />

providing the most appropriate treatments, interventions, support and services<br />

avoiding waste, including waste of equipment, supplies, ideas and energy<br />

providing care that does not vary in quality because of personal characteristics such as gender, ethnicity,<br />

geographic location or socio-economic status<br />

reducing waits and sometimes harmful and distressing delays for both those who receive care and those who<br />

give care<br />

providing care that is responsive to individual preferences, needs and values and assuring that patient values<br />

guide all clinical/health related decisions<br />

<strong>SNBTS</strong>_RDI_<strong>Strategy</strong>_2012-17_10 July2012 final.doc Page 74 of 74

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!