Become a member

#CGDSOCIETY

Gene Therapy Grants

The main objective of the CGD Society and its predecessor, the CGD Research Trust, has been to help develop a cure for CGD using gene therapy. We have invested over £2.13 million in pursuit of this goal.

Funding, which started in 1998 with proof of concept studies, has resulted in 40 publications. It has also led to commercial interest and investment in further refining the gene therapy tools that CGD Society-funded research have helped to develop. We are immensely proud that our investment in this area of research has paid off so that people with CGD can be offered gene therapy.

"Gene therapy is the start of a new branch of medicine for the treatment of inherited disorders such as CGD, and the CGD Society’s support has been invaluable in driving this field forward." Professor Len Seymour, Professor of Gene Therapies at Oxford University and Ex-president of the British Society for Gene and Cell Therapy

Research never stands still and so we have funded research into gene-editing technologies. These technologies, although in their infancy, may provide bespoke, tailored correction of the single gene defects that cause CGD.

Here we summarise the grants we have funded and their outcomes.

Tailored genetic correction of p47phox CGD

Grant awarded to: Professor Janine Reichenbach and Dr Ulrich Siler, University Children’s Hospital, Zurich, Switzerland

Amount: £50,000 over one year ending in 2017

Official title: ‘Targeted genome editing for p47phox-deficient CGD’

Aim: The p47phox form of CGD is the second most common type of CGD, with most patients carrying the same genetic mutation. This makes p47phox deficiency an attractive target for gene editing using CRISPR and TALEN technologies, which act as gene scissors. The project aimed to produce a human cell model of p47phox-deficient CGD to test the ability of CRISPR and TALEN to repair the genetic defect.

Outcomes and benefits: A cell model of p47phox deficiency was developed and validated. Comparative studies were done on the ability of CRISPR and TALEN gene tools to correct the genetic defect, and analysis was made to investigate any ‘off-target’ effects in order to better understand the safety profile of these technologies.

These essential studies will lead to pre-clinical studies in mouse models and then to the development of clinical trials.

Publications resulting from this work:

CRISPR/Cas9-generated p47phox-deficient cell line for chronic granulomatous disease gene therapy vector development
Wrona D, Siler U, Reichenbach J.
Scientific Reports, 2017 Mar 13; 7: 44187.
https://www.ncbi.nlm.nih.gov/pubmed/28287132

 

Using gene scissors to correct X-CGD

Grant awarded to: Dr Linzhao Cheng, Dr Harry Malech and Dr Jizhong Zou, Johns Hopkins University School of Medicine, Baltimore, and Laboratory of Host Defences, National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA

Amount: £50,000 over one year ending in 2011

Official title: ‘Developing novel gene therapy of X-CGD by targeted correction in patient-specific induced pluripotent stem cells’

Aim: Current gene therapy techniques rely on the use of viruses to carry a corrective healthy gene into the genetic makeup (DNA) of a patient’s cells. However, the precise location of where the healthy gene is inserted into the cell’s DNA is a random process and can lead to unwanted side effects in the gene therapy procedure. The aim was to develop a more precise method for performing gene therapy that does not use viruses for gene correction.

Outcomes and benefits: Bone marrow stem cells of a patient with X-CGD were genetically reprogrammed to become induced pluripotent stem cells (iPS cells). A specifically designed gene scissor, called a zinc finger nuclease (ZFN), was used to insert the gene gp91phox into a precise and exact location in the DNA of the patient-derived iPS cells to correct the mutation causing X-linked CGD. These corrected cells, once allowed to develop into neutrophils, were found to be able to produce superoxide like normal healthy cells.

This technology has potential advantages over virally delivered gene therapy in that it is more highly targeted and specific. This approach to gene therapy offers the potential of treating all forms of CGD regardless of the mutation involved. This is the first study that has used ZFNs in specifically targeted gene transfer to correct X-CGD. 

Publications resulting from this work:

Oxidase-deficient neutrophils from X-linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease-mediated safe harbor targeting
Zou J, Sweeney CL, Chou BK, Choi U, Pan J, Wang H, Dowey SN, Cheng L, Malech HL. Blood, 2011 May 26; 117(21): 5561–72.
http://www.ncbi.nlm.nih.gov/pubmed/21411759

 

Bringing improved lentiviral gene therapy to clinic to treat X-CGD

Grant awarded to: Professor Adrian Thrasher and Professor Christine Kinnon, Institute of Child Health, London

Amount: £308,939 over three years ending in 2011

Official title: ‘Improving gene therapy for CGD: Vector development, testing and clinical application: Part 2’

Aim: To monitor the clinical effectiveness of gene therapy in clinical trials and to use the information gained to develop new-improved gene therapy tools to treat more people with CGD.

Outcomes and benefits: Further refinements were made to the design of the lentiviral gene therapy vector to improve effectiveness and safety. The new vector was approved by the regulatory authorities and used in clinical trials in 2011. Valuable new information was gained through the clinical trials, leading to the development of a new gene vector for use in the next phase of clinical trials for not only CGD but also for X-SCID.

Work has started on testing a new wave of reagents based on an entirely different vector design. The project ensured that gene therapy could be given to those people in urgent need of this life-saving treatment.

Publications resulting from this work:

Biochemical correction of X-CGD by a novel chimeric promoter regulating high levels of transgene expression in myeloid cells
Santilli G, Almarza E, Brendel C, Choi U, Blundell MP, Haria S, Parsley KL, Kinnon C, Malech HL, Bueren JA, Grez M, Thrasher AJ.
Molecular Therapy, 2011 Jan; 19(1): 122–32.
https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3017453/

Correction of SCID-X1 using an enhancerless Vav promoter
Almarza E, Zhang F, Santilli G, Blundell MP, Howe SJ, Thornhill SI, Bueren JA, Thrasher AJ.
Human Gene Therapy, 2011 Mar; 22(3): 263–70.
http://www.ncbi.nlm.nih.gov/pubmed/20887212

Gene therapy of chronic granulomatous disease: the engraftment dilemma
Grez M, Reichenbach J, Schwäble J, Seger R, Dinauer MC, Thrasher AJ.
Molecular Therapy, 2011 Jan; 19(1): 28–35.
http://www.ncbi.nlm.nih.gov/pubmed/21045810

 

Bringing improved gene therapy to clinic for X-CGD

Grant awarded to: Dr Manuel Grez (in collaboration with Professor Adrian Thrasher), Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt, Germany

Amount: £217,703 over two years ending in 2010

Official title: ‘Improving gene therapy for CGD: Vector development, testing and clinical application: Part 2 – Development of an improved gamma retroviral vector’

Aim: Gene therapy for X-CGD over the previous six years helped save lives and showed clear clinical benefit in resolving life-threatening infections. However, 4 out of 13 people treated developed serious, severe complications after treatment. Evidence has shown that these complications were related to the design of the gene therapy reagent used. Funding supported work to determine the safety profile of a newly designed gene therapy tool called SINfes.gp91s retroviral vector.

Outcomes and benefits: The effectiveness and safety profile of the SINfes.gp91s retroviral vector was determined to give the essential pre-clinical data for the regulatory authorities. The data was submitted to regulatory authorities in Germany as part of an application for clinical trials to start in 2012. This work was essential prior to the retroviral vector’s use to treat patients with X-CGD.

Publications resulting from this work:

Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease
Stein S, Ott MG, Schultze-Strasser S, Jauch A, Burwinkel B, Kinner A, Schmidt M, Krämer A, Schwäble J, Glimm H, Koehl U, Preiss C, Ball C, Martin H, Göhring G, Schwarzwaelder K, Hofmann WK, Karakaya K, Tchatchou S, Yang R, Reinecke P, Kühlcke K, Schlegelberger B, Thrasher AJ, Hoelzer D, Seger R, von Kalle C, Grez M.
Nature Medicine, 2010 Feb; 16(2): 198–204.
http://www.ncbi.nlm.nih.gov/pubmed/20098431

A new PG13-based packaging cell line for stable production of clinical-grade self-inactivating gamma-retroviral vectors using targeted integration
Loew R, Meyer Y, Kuehlcke K, Gama-Norton L, Wirth D, Hauser H, Stein S, Grez M, Thornhill S, Thrasher A, Baum C, Schambach A.
Gene Therapy, 2010 Feb; 17(2): 272–80.
http://www.ncbi.nlm.nih.gov/pubmed/19865181

Gene therapy of chronic granulomatous disease: the engraftment dilemma
Grez M, Reichenbach J, Schwäble J, Seger R, Dinauer MC, Thrasher AJ.
Molecular Therapy, 2011 Jan; 19(1): 28–35.
http://www.ncbi.nlm.nih.gov/pubmed/21045810

 

Developing lentiviral gene medicine for X-CGD

Grant awarded to: Professor Adrian Thrasher and Professor Christine Kinnon, Molecular Immunology Unit, UCL Institute of Child Health, London

Amount: £210,864 over four years ending in 2008

Official title: ‘Improving gene therapy for CGD: Vector development, testing and clinical application: Part 1 – Development of a lentiviral vector for X-CGD’

Aim: Gene therapy using lentiviral tools is considered to be more efficient and safer than using retroviruses in gene correction. Gene therapy medicine based on a lentiviral design has been used to treat other immunodeficiency conditions, such as X-SCID. The aim of this project was to develop a new-improved lentiviral-based gene therapy reagent to treat patients with X-CGD.

Outcomes and benefits: A new gene therapy reagent was designed incorporating new safety features and two elements that ensure the corrective gene is only expressed in the cell types affected in CGD, through a process known as tissue-specific expression. This new reagent underwent further safety and effectiveness tests and was used to treat an X-CGD patient on a compassionate basis.

Publications resulting from this work:

Gene therapy of inherited immunodeficiencies
Santilli G, Thornhill SI, Kinnon C, Thrasher AJ.
Expert Opinion on Biological Therapy, 2008 Apr: 8(4): 397–407.

 

Development of a vaccine against fungal infections

Grant awarded to: Dr Brahm Segal and Dr John Subjeck, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA

Amount: £97,000 over two years ending in 2008

Official title: ‘Development of an Aspergillus vaccine’

Aim: Infection caused by the fungi Aspergillus is a serious cause of illness and mortality in CGD and in many other diseases affecting the immune system. The project aimed to develop a vaccine against Aspergillus and intrinsic to this was the further understanding of how the enzyme affected in CGD (NADPH oxidase) regulates inflammation.

Outcomes and benefits: The strategic development of the vaccine was found to be crucially dependent on understanding more about how the defect in CGD influences interactions between different types of immune cells and how this causes inflammation. The project produced a vaccine candidate and identified potential therapeutic targets, such as Nrf2 activation, for dampening down excessive inflammation in CGD.

Overall, this project gave a greater insight into how to better design immune-based therapies, including vaccination.

The work funded by the CGD Society generated preliminary results that formed the basis for a five-year $2.2m award from the National Institutes of Health to Dr Segal to continue his work on the role of NADPH oxidase in regulating inflammation. This work will be centrally relevant to CGD, but it is also broadly important to other disorders of inflammation, such as autoimmune disorders and cancer.

"Without support from the CGD Society, we would not have been able to generate the preliminary results that led to this grant being funded." Dr Segal

Publications resulting from this work:

NADPH oxidase limits innate immune responses in the lungs in mice
Segal BH, Han W, Bushey JJ, Joo M, Bhatti Z, Feminella J, Dennis CG, Vethanayagam RR, Yull FE, Capitano M, Wallace PK, Minderman H, Christman JW, Sporn MB, Chan J, Vinh DC, Holland SM, Romani LR, Gaffen SL, Freeman ML, Blackwell TS.
PLoS One, 2010 Mar 16; 5(3): e9631.
http://www.plosone.org/article/info%3Adoi%2F10.1371%2Fjournal.pone.0009631

 

Developing an improved gene therapy reagent to treat X-CGD

Grant awarded to: Dr Manuel Grez (in collaboration with Professor Adrian Thrasher), Department of Applied Virology and Gene Therapy, Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt, Germany

Amount: £310,227 over three years ending in 2008

Official title: ‘Improving gene therapy for CGD: Vector development, testing and clinical application: Development of an improved gamma retroviral vector’

Aim: This project built on information gained from clinical trials for X-linked CGD, using retroviral-based gene therapy reagent, indicating that changes in design were needed to improve safety and effectiveness.

Outcomes and benefits: A safer retroviral reagent called SINfes.gp91s was developed. Major developments were the incorporation of a DNA sequence that drives the expression of the corrective gene gp91phox such that it will only be expressed in phagocytes, and optimisation of the corrective gene sequence so that it works more efficiently.

Publications resulting from this work:

Gene therapy of chronic granulomatous disease: the engraftment dilemma
Grez M, Reichenbach J, Schwäble J, Seger R, Dinauer MC, Thrasher AJ.
Molecular Therapy, 2011 Jan; 19(1): 28–35.
http://www.ncbi.nlm.nih.gov/pubmed/21045810

Advances in the treatment of chronic granulomatous disease by gene therapy
Ott MG, Seger R, Stein S, Siler U, Hoelzer D, Grez M.
Current Gene Therapy, 2007 Jun; 7(3): 155–61.

http://www.ncbi.nlm.nih.gov/pubmed/17584034

Progress and prospects: gene therapy clinical trials (part 1)
Alexander BL, Ali RR, Alton EW, Bainbridge JW, Braun S, Cheng SH, Flotte TR, Gaspar HB, Grez M, Griesenbach U, Kaplitt MG, Ott MG, Seger R, Simons M, Thrasher AJ, Thrasher AZ, Ylä-Herttuala S.
Gene Therapy, 2007 Oct; 14(20): 1439–47. Review. Erratum in: Gene Therapy, 2007 Dec; 14(24): 1754.
http://www.ncbi.nlm.nih.gov/pubmed/17909539

Transgene optimization significantly improves SIN vector titers, gp91phox expression and reconstitution of superoxide production in X-CGD cells
Moreno-Carranza B, Gentsch M, Stein S, Schambach A, Santilli G, Rudolf E, Ryser MF, Haria S, Thrasher AJ, Baum C, Brenner S, Grez M.
Gene Therapy, 2009, 16: 111–118.
http://www.ncbi.nlm.nih.gov/pubmed/18784749

 

Testing improved gene therapy vectors

Grant awarded to: Professor Mary Dinauer, Herman B Wells Centre for Paediatric Research, Indiana University School of Medicine, USA

Amount: $20,000 over one year ending in 2008; made possible through a generous donation from the Chronic Granulomatous Disease Association, USA

Official title: ‘Testing improved gene therapy vectors for treatment of infection and chronic inflammation in CGD’

Aim: The research examined how effective a new gene therapy tool, known as SINfes.gp91s, developed by Dr Manuel Grez and colleagues, was at restoring the ability to fight infections and resolve inflammation in a mouse model of X-CGD.

Outcomes and benefits: New systems were developed to test the effectiveness of gene therapy in fighting different types of infection in CGD. The work determined how much gene correction is necessary to protect against infection. Survival experiments using Aspergillus fumigatus, Burkholderia cepacia and Staphylococcus aureus showed that different levels of oxidase correction are needed to stave off different infections. Levels needed ranged from 10% to 25% gene-corrected cells.

 

Gene therapy clinical trials for X-CGD

Grant awarded to: Professor Adrian Thrasher, Dr Manuel Grez and Dr Hanspeter Hossle, Institute of Child Health, University College London, Georg-Speyer-Haus, Frankfurt, Germany and the University Children’s Hospital, Zurich, Switzerland

Amount: £106,000 over two years ending in 2004

Official title: ‘Comparative gene therapy trial for X-CGD at three European centres’

Aim: This proposal funded the production of clinical grade gene therapy vector, known as SF71gp91phox, for the treatment of X-CGD patients by gene therapy. 

Outcomes and benefits: The reagent was used to treat seven X-CGD patients (four children and three adults) in clinical trials for CGD in London, Frankfurt and Zurich. The evidence from the trials showed that gene therapy has the potential to provide significant clinical benefit for CGD patients in the short term but can have significant safety risks. Post-gene therapy benefits included clearing of life-threatening infections that were resistant to treatment by conventional medicine. However, three patients, treated in Zurich and Frankfurt, reported severe toxic side effects of treatment, including developing a form of leukaemia.

The results showed that the effectiveness of gene therapy was only transitory, with levels of gene-corrected cells dropping substantially within months of treatment. This is known to be due to ‘silencing’ of the corrective gene and the corrected cells not fully engrafting within the bone marrow.

Valuable insight was gained about the problems that need to be overcome to make gene therapy a safe and more effective treatment, and potentially a cure, for CGD. As a consequence, the CGD Society set aside £1 million in funding for subsequent projects to develop new regents to treat X-CGD.  

Publications resulting from this work:

Gene therapy of chronic granulomatous disease: the engraftment dilemma
Grez M, Reichenbach J, Schwäble J, Seger R, Dinauer MC, Thrasher AJ.
Molecular Therapy, 2011 Jan; 19(1): 28–35. Review.
http://www.ncbi.nlm.nih.gov/pubmed/21045810

Advances in the treatment of chronic granulomatous disease by gene therapy
Ott MG, Seger R, Stein S, Siler U, Hoelzer D, Grez M.
Current Gene Therapy, 2007 Jun; 7(3):155–61. Review.

http://www.ncbi.nlm.nih.gov/pubmed/17584034

Progress and prospects: gene therapy clinical trials (part 1)
Alexander BL, Ali RR, Alton EW, Bainbridge JW, Braun S, Cheng SH, Flotte TR, Gaspar HB, Grez M, Griesenbach U, Kaplitt MG, Ott MG, Seger R, Simons M, Thrasher AJ, Thrasher AZ, Ylä-Herttuala S.
Gene Therapy, 2007 Oct; 14(20): 1439–47. Review. Erratum in: Gene Therapy, 2007 Dec; 14(24): 1754.
http://www.ncbi.nlm.nih.gov/pubmed/17909539

 

Using protein transfer to treat X-CGD

Grant awarded to: Professor Françoise Morel, Dr Marie-Hélène Paclet and Dr Jean-Luc Lenormand, Groupe de Recherche et d’Etude du Processus Inflammatoire, University Joseph Fourier, Grenoble, France

Amount: £30,083 over one year ending in 2007

Official title: ‘Protein transfer as a new therapeutic approach in X-linked CGD’

Aim: To explore a new alternative treatment for CGD by putting the protein that is missing in CGD into cells rather than the gene.

Outcomes and benefits: This study established proof of principle, showing that using special delivery agents called proteoliposomes, Gp91phox, the protein affected in X-linked CGD, can be transferred into cells. Once taken up by the cells, the artificial Gp91phox showed similar activities to those associated with its natural counterpart.

The work showed that protein therapy could be another option for the treatment of CGD in the future. This therapy could help ‘top up’ patients’ immune systems over a short period of time to fight off infections.

Publications resulting from this work:

Liposome-mediated cellular delivery of active gp91phox
Marques B, Liguori L, Paclet M-H, Villegas-Mendéz A, Rothe R, Morel F, Lenormand JL. PLoS One, 2007 Sep 12; 2(9): e856.

www.plosone.org/article/info%3Adoi%2F10.1371%2Fjournal.pone.0000856

New insights into the membrane topology of the phagocyte NADPH oxidase: characterization of an anti-gp91-phox conformational monoclonal antibody
Campion Y, Paclet MH, Jesaitis AJ, Marques B, Grichine A, Berthier S, Lenormand JL, Lardy B, Stasia MJ, Morel F.
Biochimie, 2007 Sep; 89(9): 1145–58.
http://www.ncbi.nlm.nih.gov/pubmed/17397983

 

Studying how the immune system recovers after gene therapy

Grant awarded to: Professor Adrian Thrasher, Professor Bobby Gaspar and Professor Christine Kinnon, Institute of Child Health, University College London

Amount: £110,259 over two years ending in 2005

Official title: ‘Analysis of immune reconstitution and function in clinical trials of gene therapy for CGD’

Aim: This project supported ongoing gene therapy clinical trials for patients with X-linked CGD and other primary immunodeficiency diseases.

Outcomes and benefits: Methods were developed to measure how many copies of corrected genes are delivered into patients’ cells after gene therapy and exactly where, at the DNA level, the healthy gene is integrated and in what cell types. Improvements were made to the design of gene therapy tools for the treatment of CGD.

The project funded facilities for the storage and cataloguing of patients’ samples so that measurement and monitoring of corrected gene levels can be done over long periods of time for people undergoing gene therapy for a number of genetic disorders. This important project helped safer gene therapy procedures to be developed.

Publications resulting from this work:

Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector
Gaspar HB, Parsley KL, Howe S, King D, Gilmour KC, Sinclair J, Brouns G, Schmidt M, Von Kalle C, Barington T, Jakobsen MA, Christensen HO, Al Ghonaium A, White HN, Smith JL, Levinsky RJ, Ali RR, Kinnon C, Thrasher AJ.
Lancet, 2004 Dec 18-31; 364(9452): 2181–7.

http://www.ncbi.nlm.nih.gov/pubmed/15610804

Failure of SCID-X1 gene therapy in older patients
Thrasher AJ, Hacein-Bey-Abina S, Gaspar HB, Blanche S, Davies EG, Parsley K, Gilmour K, King D, Howe S, Sinclair J, Hue C, Carlier F, von Kalle C, de Saint Basile G, le Deist F, Fischer A, Cavazzana-Calvo, M.
Blood, 2005 Jun 1; 105(11): 4255–7.

http://bloodjournal.hematologylibrary.org/content/105/11/4255.long

Lentiviral vectors containing an enhancer-less ubiquitously acting chromatin opening element (UCOE) provide highly reproducible and stable transgene expression in haematopoietic cells
Zhang F, Thornhill SI, Howe SJ, Ulaganathan M, Schambach A, Sinclair J, Kinnon C, Gaspar HB, Antoniou M, Thrasher AJ.
Blood, 2007 Sep 1; 110(5): 1448–57.
http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2629730&tool+pmcentrez

 

New strategies for the treatment of CGD

Grant awarded to: Professors Adrian Thrasher, David Goldblatt and Christine Kinnon, Institute of Child Health, University College London

Amount: £190,291 over four years ending in 2004

Official title: ‘Therapeutic strategies for the treatment of CGD: Development of a clinical gene therapy protocol for CGD’

Aim: The aim of this grant award was to start gene therapy trials for the treatment of CGD.

Outcomes and benefits: During the lifetime of the grant a state-of-the art clinical gene therapy facility was established at Great Ormond Street Hospital. Protocols and standard operating procedures were put in place for three Phase 1 clinical studies following approval from the Gene Therapy Advisory Committee. Nine patients – seven with X-linked severe combined immunodeficiency (SCID-X1), one with adenosine deaminase deficiency and one with X-CGD – received gene therapy.

Highly efficient gene transfer tools based on a lentiviral design were developed for the treatment of X-CGD. The CGD Society grant also helped to fund pioneering work on the treatment of inherited eye diseases by gene therapy, using this type of gene therapy transfer tool. This work continues to benefit gene therapy trials for the treatment of many disorders.

Publications resulting from this work:

High-level transduction and gene expression in hematopoietic repopulating cells using a human immunodeficiency [correction of immunodeficiency] virus type 1-based lentiviral vector containing an internal spleen focus forming virus promoter
Demaison C, Parsley K, Brouns G, Scherr M, Battmer K, Kinnon C, Grez M, Thrasher AJ. Human Gene Therapy, 2002 May 1; 13(7): 803–13.
http://www.ncbi.nlm.nih.gov/pubmed/11975847

Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector
Gaspar HB, Parsley KL, Howe S, King D, Gilmour KC, Sinclair J, Brouns G, Schmidt M, Von Kalle C, Barington T, Jakobsen MA, Christensen HO, Al Ghonaium A, White HN, Smith JL, Levinsky RJ, Ali RR, Kinnon C, Thrasher AJ.
Lancet, 2004 Dec 18-31; 364(9452): 2181–7.
http://www.ncbi.nlm.nih.gov/pubmed/15610804

In vivo gene transfer to the mouse eye using an HIV-based lentiviral vector; efficient long-term transduction of corneal endothelium and retinal pigment epithelium
Bainbridge JW, Stephens C, Parsley K, Demaison C, Halfyard A, Thrasher AJ, Ali RR. Gene Therapy, 2001 Nov; 8(21): 1665–8.
http://www.nature.com/gt/journal/v8/n21/full/3301574a.html

 

Increasing the chances of success for CGD gene therapy

Grant awarded to: Dr Manuel Grez, Institute for Biomedical Research, Georg-Speyer-Haus, Frankfurt, Germany, in collaboration with Institute of Child Health, London

Amount: £182,961 over three years ending in 2004

Official title: ‘Development of strategies for the selective expansion of transduced CGD cells’

Aim: To explore strategies to increase the success of gene therapy.

Outcomes and benefits: In CGD, the gene-corrected cells do not have any natural selective advantage over the non-corrected cells present in the bone marrow. This project helped to set up methods to increase the number of cells carrying the corrective gene gp91phox and increase their survival once transplanted back into patients.

Publications resulting from this work:

Mobilization and transduction of CD34(+) peripheral blood stem cells in patients with X-linked chronic granulomatous disease
Ott MG, Merget-Millitzer H, Ottmann OG, Martin H, Brüggenolte N, Bialek H, Seger R, Hossle JP, Hoelzer D, Grez M.
Journal of Hematotherapy and Stem Cell Research, 2002 Aug; 11(4): 683–94.
http://www.ncbi.nlm.nih.gov/pubmed/12201957

 

Transferring therapeutic genes in CGD

Grant awarded to: Professor JP Hossle and Professor R Seger, University Children’s Hospital, Zurich, Switzerland

Amount: £84,401 over two years ending in 2002

Official title: ‘Natural tissue-specific gene regulation after therapeutic gene transfer in CGD’

Aim: To refine gene therapy for X-CGD so that the corrective gene, gp91phox, is only expressed in specific types of blood cells.

Outcomes and benefits: Two important gene sequences capable of regulating gp91phox were identified. The work added to the process of developing better gene therapy tools for X-CGD.

Publications resulting from this work:

Gene therapy of hematopoietic stem cells: Strategies for improvement
Hossle JP, Seger RA, Steinhoff D.
News in Physiological Sciences, 2002 17: 87–92.
http://physiologyonline.physiology.org/content/17/3/87.long

Lentivirus-mediated gene transfer of gp91phox corrects chronic granulomatous disease (CGD) phenotype in human X-CGD cells
Saulnier SO, Steinhoff D, Dinauer MC, Zufferey R, Trono D, Seger RA, Hossle JP.
Journal of Gene Medicine, 2000 Sep–Oct; 2(5): 317–25.
http://www.ncbi.nlm.nih.gov/pubmed/11045425

 

Learning more about the p47phox gene

Grant awarded to: Dr Colin Casimir, Department of Haematology, Imperial College London

Amount: £81,475 over two years starting in 2001

Official title: ‘Factors controlling the myeloid cell specific regulation of the p47phox gene’

Aim: The p47phox protein is missing in about one quarter to one third of all CGD patients and is the most common cause of autosomal recessive CGD (the type that affects boys and girls equally). This project investigated the gene that codes for p47phox, in particular, how the gene is regulated, so that it is highly active in phagocytes but not in other types of cells.

Outcomes and benefits: The work identified two regions of DNA that are important for p47phox gene expression. This knowledge about the regulation of the p47phox gene was used to design better and more effective gene therapy vectors with the potential for treating CGD.

Publications resulting from this work:

Enhancer-deleted retroviral vectors restore high levels of superoxide generation in a mouse model of CGD
Schwickerath O, Brouns G, Thrasher A, Kinnon C, Roes J, Casimir C.
Journal of Gene Medicine, 2004 Jun; 6(6): 603–15.

http://www.ncbi.nlm.nih.gov/pubmed/15170731

A functional ISRE is required for myeloid transcription of the p47(phox) gene
Marden C, Cunninghame Graham D, Thrasher A, Casimir C.
Biochimica et Biophysica Acta, 2003 Nov 30; 1630(2–3): 117–22. http://www.ncbi.nlm.nih.gov/pubmed/14654241

Differentiation-dependent up-regulation of p47(phox) gene transcription is associated with changes in PU.1 phosphorylation and increased binding affinity
Marden CM, Stefanidis D, Cunninghame-Graham DS, Casimir CM.
Biochemical and Biophysical Research Communications, 2003 May 23; 305(1): 193–202.

http://www.ncbi.nlm.nih.gov/pubmed/12732216

Functional analysis of the p47phox gene promoter

Marden CM; PhD thesis, University of London (1999).

 

Improving gene therapy for CGD

Grant awarded to: Dr Adrian Thrasher, Department of Molecular Biology, Institute of Child Health, University College London

Amount: £98,000 over two years starting in 1999

Official title: ‘Investigation of alternative viruses and viral promotors that may be effective in achieving better gene therapy results’

Aim: To explore different types of gene therapy tools for use in treating genetic disorders.

Outcomes and benefits: This work helped improve gene transfer into human stem cells and showed that lentiviral vectors could be highly effective for transferring therapeutic genes into stem cells. This pioneering, proof-of-principle work paved the way for gene therapy for X-SCID and CGD.

Publications resulting from this work:

A defined window for efficient gene marking of severe combined immunodeficient-repopulating cells using a gibbon ape leukemia virus-pseudotyped retroviral vector Demaison C, Brouns G, Blundell MP, Goldman JP, Levinsky RJ, Grez M, Kinnon C, Thrasher AJ.
Human Gene Therapy, 2000 Jan 1; 11(1): 91–100.
http://www.ncbi.nlm.nih.gov/pubmed/10646642

High-level transduction and gene expression in hematopoietic repopulating cells using a human immunodeficiency [correction of immunodeficiency] virus type 1-based lentiviral vector containing an internal spleen focus forming virus promoter
Demaison C, Parsley K, Brouns G, Scherr M, Battmer K, Kinnon C, Grez M, Thrasher AJ. Human Gene Therapy, 2002 May 1; 13(7): 803–13.
http://www.ncbi.nlm.nih.gov/pubmed/11975847

 

Developing model systems to test gene therapy reagents

Grant awarded to: Dr Adrian Thrasher, Department of Molecular Biology, Institute of Child Health, University College London

Amount: £50,000 over three years starting in 1998

Official title: ‘To develop pre-clinical models for treatment of CGD by gene therapy’

Aim: Before gene therapy can be used to treat patients, it has to be tested in animal CGD models to show its effectiveness and safety.

Outcomes and benefits: Animal models representing the X-CGD and p47phox types of CGD were developed. This work was of lasting benefit as all pre-clinical work on testing gene therapy medicine for use in CGD patients uses these animal models.

Publications resulting from this work:

Chronic granulomatous disease
Goldblatt D, Thrasher AJ.
Clinical and Experimental Immunology, 2000 Oct; 122(1): 1–9. Review. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1905749/?tool=pubmed

 

Improving gene transfer into human stem cells

Grant awarded to: Dr Colin Casimir, Department of Haematology, Imperial College, London

Amount: £34,141 over one year ending in 1998Official title: ‘Factors affecting the growth and survival of pluripotent human haematopoietic cells in vitro’

Aim: Putting a corrective gene into stem cells is a challenge in gene therapy. This project investigated ways of transferring genes effectively into human stem cells.

Outcomes and benefits: A new method was developed that allowed the transfer of genes into stem cells from cord blood.

Publications resulting from this work:

Retroviral transduction of quiescent haematopoietic cells using a packaging cell line expressing the membrane-bound form of stem cell factor
Sehgal A, Weeratunge N, Casimir C.
Gene Therapy 6, 1999; 1084–91.
http://www.nature.com/gt/journal/v6/n6/full/3300932a.html

Enhanced retroviral transduction of 5-fluorouracil-resistant human bone marrow (stem) cells using a genetically modified packaging cell line
Povey J, Weeratunge N, Marden C, Sehgal A, Thrasher A, Casimir C.
Blood, 1998; 92: 4080–9.
http://bloodjournal.hematologylibrary.org/content/92/11/4080.long

Other publications resulting from CGD Society funding prior to 1997

High efficiency gene transfer to human hematopoietic SCID-repopulating cells under serum-free conditions
Schilz AJ, Brouns G, Knöss H, Ottmann OG, Hoelzer D, Fauser AA, Thrasher AJ, Grez M. Blood, 1998; 92: 3163–71.
http://bloodjournal.hematologylibrary.org/content/92/9/3163.long

Gene transfer to primary chronic granulomatous disease monocytes
Thrasher AJ, Casimir CM, Kinnon C, Morgan G, Segal AW, Levinsky RJ.
Lancet, 1995 Jul 8; 346(8967): 92–3.
http://www.ncbi.nlm.nih.gov/pubmed/7541496

Functional reconstitution of the NADPH-oxidase by adeno-associated virus gene transfer.
Thrasher AJ, de Alwis M, Casimir CM, Kinnon C, Page K, Lebkowski J, Segal AW, Levinsky RJ.
Blood. 1995; 15;86(2):761- 5.
http://www.bloodjournal.org/content/86/2/761.long?sso-checked=true

Generation of recombinant adeno-associated virus (rAAV) from an adenoviral vector and functional reconstitution of the NADPH-oxidase
Thrasher AJ, de Alwis M, Casimir CM, Kinnon C, Page K, Lebkowski J, Segal AW, Levinsky RJ.
Gene Therapy, 1995 Sep; 2(7): 481–5.
http://www.ncbi.nlm.nih.gov/pubmed/7584126

 

More information

Our website contains a wealth of information to help and support you. If you are not able to find the answer to a specific question, feel free to contact us using the form at the bottom of the page or by emailing or calling us.  We are here to help.