201
|
Targeted Gene Correction in Osteopetrotic-Induced Pluripotent Stem Cells for the Generation of Functional Osteoclasts. Stem Cell Reports 2015; 5:558-68. [PMID: 26344905 PMCID: PMC4624934 DOI: 10.1016/j.stemcr.2015.08.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 08/05/2015] [Accepted: 08/05/2015] [Indexed: 01/15/2023] Open
Abstract
Autosomal recessive osteopetrosis is a human bone disease mainly caused by TCIRG1 gene mutations that prevent osteoclasts resorbing activity, recapitulated by the oc/oc mouse model. Bone marrow transplantation is the only available treatment, limited by the need for a matched donor. The use of induced pluripotent stem cells (iPSCs) as an unlimited source of autologous cells to generate gene corrected osteoclasts might represent a powerful alternative. We generated iPSCs from oc/oc mice, corrected the mutation using a BAC carrying the entire Tcirg1 gene locus as a template for homologous recombination, and induced hematopoietic differentiation. Similarly to physiologic fetal hematopoiesis, iPSC-derived CD41+ cells gradually gave rise to CD45+ cells, which comprised both mature myeloid cells and high proliferative potential colony-forming cells. Finally, we differentiated the gene corrected iPSC-derived myeloid cells into osteoclasts with rescued bone resorbing activity. These results are promising for a future translation into the human clinical setting. iPSCs from oc/oc mice bearing Tcirg1 gene mutation were generated for the first time A BAC-based approach corrects the Tcirg1 gene mutation iPSCs differentiate similarly to physiologic fetal hematopoiesis The osteopetrotic phenotype in osteoclasts from BAC-corrected iPSCs was rescued
Collapse
|
202
|
Unguru Y. Ethical Challenges in Early-Phase Pediatric Research for Life-Limiting Illness. Semin Pediatr Neurol 2015; 22:177-86. [PMID: 26358428 DOI: 10.1016/j.spen.2015.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Research with children is necessary to assure more effective treatments and potential cures of childhood illnesses. Ethical conduct of research requires minimizing the inherent risks of research, especially when it involves vulnerable populations such as children. Duchenne muscular dystrophy (DMD) is a progressive and fatal disease with no Food and Drug Administration-approved treatment. Clinical trials investigating so-called gene therapies are viewed by many in the DMD community with great promise. The goal of research is to secure generalizable knowledge and not directly benefit patients, yet many parents of boys with DMD hope, and even expect, that their sons will derive medical benefit by participating in early-phase "gene therapy" trials, raising concern for the therapeutic misconception. Physician-investigators must assist patient-subjects to distinguish realistic from unrealistic hope while maintaining reasonable expectations. In this article, I examine the therapeutic misconception and related concepts as framed within the context of "gene therapy" for DMD.
Collapse
Affiliation(s)
- Yoram Unguru
- Division of Pediatric Hematology/Oncology, The Herman and Walter Samuelson Children's Hospital at Sinai, Baltimore, MD; Johns Hopkins University, Berman Institute of Bioethics, Baltimore, MD.
| |
Collapse
|
203
|
Tai DS, Hu C, Lee CCI, Martinez M, Cantero G, Kim EH, Tarantal AF, Lipshutz GS. Development of operational immunologic tolerance with neonatal gene transfer in nonhuman primates: preliminary studies. Gene Ther 2015; 22:923-30. [PMID: 26333349 DOI: 10.1038/gt.2015.65] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 05/22/2015] [Accepted: 06/18/2015] [Indexed: 02/05/2023]
Abstract
Achieving persistent expression is a prerequisite for effective genetic therapies for inherited disorders. These proof-of-concept studies focused on adeno-associated virus (AAV) administration to newborn monkeys. Serotype rh10 AAV expressing ovalbumin and green fluorescent protein (GFP) was administered intravenously at birth and compared with vehicle controls. At 4 months postnatal age, a second injection was administered intramuscularly, followed by vaccination at 1 year of age with ovalbumin and GFP. Ovalbumin was highest 2 weeks post administration in the treated monkey, which declined but remained detectable thereafter; controls demonstrated no expression. Long-term AAV genome copies were present in myocytes. At 4 weeks, neutralizing antibodies to rh10 were present in the experimental animal only. With AAV9 administration at 4 months, controls showed transient ovalbumin expression that disappeared with the development of strong anti-ovalbumin and anti-GFP antibodies. In contrast, increased and maintained ovalbumin expression was noted in the monkey administered AAV at birth, without antibody development. After vaccination, the experimental monkey maintained levels of ovalbumin without antibodies, whereas controls demonstrated high levels of antibodies. These preliminary studies suggest that newborn AAV administration expressing secreted and intracellular xenogenic proteins may result in persistent expression in muscle, and subsequent vector administration can result in augmented expression without humoral immune responses.
Collapse
Affiliation(s)
- D S Tai
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - C Hu
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - C C I Lee
- California National Primate Research Center and Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - M Martinez
- California National Primate Research Center and Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - G Cantero
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - E H Kim
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - A F Tarantal
- California National Primate Research Center and Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| | - G S Lipshutz
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
204
|
Abstract
In this issue of Blood, Touzot et al report that autologous gene therapy/hematopoietic stem cell transplantation (HSCT) for infants with X-linked severe combined immune deficiency (SCID-X1) lacking a matched sibling donor may have better outcomes than haploidentical (haplo) HSCT. Because gene therapy represents an autologous transplant, it obviates immune suppression before and after transplant, eliminates risks of graft versus host disease (GVHD), and, as the authors report, led to faster immunological reconstitution after transplant than did haplo transplant.
Collapse
|
205
|
Abstract
The thymus is an essential organ for the generation of the adaptive immune system. By now, the cellular selection events taking place in ongoing life before sexual maturity have been worked out even at the molecular level, and thus thymic lymphocyte development represents one of the best-studied systems in mammalian development. Because thymic lymphocyte development involves ample proliferation and generation of new cells, it is not astonishing that the thymus also represents an organ where malignancy can develop. In this Masters of Immunology primer, the development of lymphocytes and the role of intracellular Notch 1 and cyclins in lymphocytic malignancy are reviewed, offering new therapeutic possibilities.
Collapse
Affiliation(s)
- Harald von Boehmer
- Author's Affiliations: Harvard Medical School; Dana-Farber Cancer Institute, Boston, Massachusetts; University of Florida, Gainesville, Florida; and University of Munich, Munich, Germany
| |
Collapse
|
206
|
Hashemi M, Ayatollahi S, Parhiz H, Mokhtarzadeh A, Javidi S, Ramezani M. PEGylation of Polypropylenimine Dendrimer with Alkylcarboxylate Chain Linkage to Improve DNA Delivery and Cytotoxicity. Appl Biochem Biotechnol 2015; 177:1-17. [PMID: 26162520 DOI: 10.1007/s12010-015-1723-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/22/2015] [Indexed: 12/11/2022]
|
207
|
Worth AJJ, Thrasher AJ. Current and emerging treatment options for Wiskott–Aldrich syndrome. Expert Rev Clin Immunol 2015; 11:1015-32. [DOI: 10.1586/1744666x.2015.1062366] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
208
|
Vannocci T, Faggianelli N, Zaccagnino S, della Rosa I, Adinolfi S, Pastore A. A new cellular model to follow Friedreich's ataxia development in a time-resolved way. Dis Model Mech 2015; 8:711-9. [PMID: 26035392 PMCID: PMC4486863 DOI: 10.1242/dmm.020545] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/20/2015] [Indexed: 01/30/2023] Open
Abstract
Friedreich's ataxia (FRDA) is a recessive autosomal ataxia caused by reduced levels of frataxin (FXN), an essential mitochondrial protein that is highly conserved from bacteria to primates. The exact role of frataxin and its primary function remain unclear although this information would be very valuable to design a therapeutic approach for FRDA. A main difficulty encountered so far has been that of establishing a clear temporal relationship between the different observations that could allow a distinction between causes and secondary effects, and provide a clear link between aging and disease development. To approach this problem, we developed a cellular model in which we can switch off/on in a time-controlled way the frataxin gene partially mimicking what happens in the disease. We exploited the TALEN and CRISPR methodologies to engineer a cell line where the presence of an exogenous, inducible FXN gene rescues the cells from the knockout of the two endogenous FXN genes. This system allows the possibility of testing the progression of disease and is a valuable tool for following the phenotype with different newly acquired markers.
Collapse
Affiliation(s)
- Tommaso Vannocci
- Molecular Structure Division, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | - Nathalie Faggianelli
- Molecular Structure Division, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | - Silvia Zaccagnino
- Molecular Structure Division, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | - Ilaria della Rosa
- Molecular Structure Division, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | - Salvatore Adinolfi
- Department of Basic and Clinical Neurosciences, Kings College London, London NW7 1AA, UK
| | - Annalisa Pastore
- Department of Basic and Clinical Neurosciences, Kings College London, London NW7 1AA, UK
| |
Collapse
|
209
|
Progress in gene therapy for primary immunodeficiencies using lentiviral vectors. Curr Opin Allergy Clin Immunol 2015; 14:527-34. [PMID: 25207699 DOI: 10.1097/aci.0000000000000114] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW This review gives an overview over the most recent progress in the field of lentiviral gene therapy for primary immunodeficiencies (PIDs). The history and state-of-the-art of lentiviral vector development are summarized and the recent advancements for a number of selected diseases are reviewed in detail. Past retroviral vector trials for these diseases, the most recent improvements of lentiviral vector platforms and their application in preclinical development as well as ongoing clinical trials are discussed. RECENT FINDINGS Main focus is on the preclinical studies and clinical trials for the treatment of Wiskott-Aldrich syndrome, chronic granulomatous disease, adenosine deaminase deficient severe combined immunodeficiency (ADA-SCID) and X-linked severe combined immunodeficiency with lentiviral gene therapy. SUMMARY Gene therapy for PIDs is an effective treatment, providing potential long-term clinical benefit for affected patients. Substantial progress has been made to make lentiviral gene therapy platforms available for a number of rare genetic diseases. Although many ongoing gene therapy trials are based on ex-vivo approaches with autologous hematopoietic stem cells, other approaches such as in-vivo gene therapy or gene-repair platforms might provide further advancement for certain PIDs.
Collapse
|
210
|
Huston MW, Riegman ARA, Yadak R, van Helsdingen Y, de Boer H, van Til NP, Wagemaker G. Pretransplant mobilization with granulocyte colony-stimulating factor improves B-cell reconstitution by lentiviral vector gene therapy in SCID-X1 mice. Hum Gene Ther 2015; 25:905-14. [PMID: 25222508 DOI: 10.1089/hum.2014.101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hematopoietic stem cell (HSC) gene therapy is a demonstrated effective treatment for X-linked severe combined immunodeficiency (SCID-X1), but B-cell reconstitution and function has been deficient in many of the gene therapy treated patients. Cytoreductive preconditioning is known to improve HSC engraftment, but in general it is not considered for SCID-X1 since the poor health of most of these patients at diagnosis and the risk of toxicity preclude the conditioning used in standard bone marrow stem cell transplantation. We hypothesized that mobilization of HSC by granulocyte colony-stimulating factor (G-CSF) should create temporary space in bone marrow niches to improve engraftment and thereby B-cell reconstitution. In the present pilot study supplementing our earlier preclinical evaluation (Huston et al., 2011), Il2rg(-/-) mice pretreated with G-CSF were transplanted with wild-type lineage negative (Lin(-)) cells or Il2rg(-/-) Lin(-) cells transduced with therapeutic IL2RG lentiviral vectors. Mice were monitored for reconstitution of lymphocyte populations, level of donor cell chimerism, and antibody responses as compared to 2 Gy total body irradiation (TBI), previously found effective in promoting B-cell reconstitution. The results demonstrate that G-CSF promotes B-cell reconstitution similar to low-dose TBI and provides proof of principle for an alternative approach to improve efficacy of gene therapy in SCID patients without adverse effects associated with cytoreductive conditioning.
Collapse
Affiliation(s)
- Marshall W Huston
- 1 Department of Neurology, Erasmus University Medical Center , 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
211
|
Guerrero AD, Moyes JS, Cooper LJN. The human application of gene therapy to re-program T-cell specificity using chimeric antigen receptors. CHINESE JOURNAL OF CANCER 2015; 33:421-33. [PMID: 25189715 PMCID: PMC4190432 DOI: 10.5732/cjc.014.10100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The adoptive transfer of T cells is a promising approach to treat cancers. Primary human T cells can be modified using viral and non-viral vectors to promote the specific targeting of cancer cells via the introduction of exogenous T-cell receptors (TCRs) or chimeric antigen receptors (CARs). This gene transfer displays the potential to increase the specificity and potency of the anticancer response while decreasing the systemic adverse effects that arise from conventional treatments that target both cancerous and healthy cells. This review highlights the generation of clinical-grade T cells expressing CARs for immunotherapy, the use of these cells to target B-cell malignancies and, particularly, the first clinical trials deploying the Sleeping Beauty gene transfer system, which engineers T cells to target CD19+ leukemia and non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Alan D Guerrero
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
212
|
Ghosh S, Thrasher AJ, Gaspar HB. Gene therapy for monogenic disorders of the bone marrow. Br J Haematol 2015; 171:155-170. [DOI: 10.1111/bjh.13520] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Sujal Ghosh
- Infection, Immunity, Inflammation and Physiological Medicine; Molecular and Cellular Immunology Section; University College London - Institute of Child Health; London UK
- Department of Paediatric Oncology, Haematology and Clinical Immunology; Medical Faculty; Centre of Child and Adolescent Health; Heinrich-Heine-University; Düsseldorf Germany
| | - Adrian J. Thrasher
- Infection, Immunity, Inflammation and Physiological Medicine; Molecular and Cellular Immunology Section; University College London - Institute of Child Health; London UK
| | - H. Bobby Gaspar
- Infection, Immunity, Inflammation and Physiological Medicine; Molecular and Cellular Immunology Section; University College London - Institute of Child Health; London UK
| |
Collapse
|
213
|
Ott de Bruin LM, Volpi S, Musunuru K. Novel Genome-Editing Tools to Model and Correct Primary Immunodeficiencies. Front Immunol 2015; 6:250. [PMID: 26052330 PMCID: PMC4440404 DOI: 10.3389/fimmu.2015.00250] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/07/2015] [Indexed: 12/22/2022] Open
Abstract
Severe combined immunodeficiency (SCID) and other severe non-SCID primary immunodeficiencies (non-SCID PID) can be treated by allogeneic hematopoietic stem cell (HSC) transplantation, but when histocompatibility leukocyte antigen-matched donors are lacking, this can be a high-risk procedure. Correcting the patient's own HSCs with gene therapy offers an attractive alternative. Gene therapies currently being used in clinical settings insert a functional copy of the entire gene by means of a viral vector. With this treatment, severe complications may result due to integration within oncogenes. A promising alternative is the use of endonucleases such as ZFNs, TALENs, and CRISPR/Cas9 to introduce a double-stranded break in the DNA and thus induce homology-directed repair. With these genome-editing tools a correct copy can be inserted in a precisely targeted "safe harbor." They can also be used to correct pathogenic mutations in situ and to develop cellular or animal models needed to study the pathogenic effects of specific genetic defects found in immunodeficient patients. This review discusses the advantages and disadvantages of these endonucleases in gene correction and modeling with an emphasis on CRISPR/Cas9, which offers the most promise due to its efficacy and versatility.
Collapse
Affiliation(s)
- Lisa M Ott de Bruin
- Division of Immunology, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Pediatric Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht , Utrecht , Netherlands
| | - Stefano Volpi
- UO Pediatria 2, Istituto Giannina Gaslini, University of Genoa , Genoa , Italy ; Division of Immunology and Allergy, Laboratory Center of Epalinges (CLE), University Hospital of Lausanne , Epalinges , Switzerland
| | - Kiran Musunuru
- Department of Stem Cell and Regenerative Biology, Harvard University , Cambridge, MA , USA
| |
Collapse
|
214
|
Li KC, Hu YC. Cartilage tissue engineering: recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater 2015; 4:948-68. [PMID: 25656682 DOI: 10.1002/adhm.201400773] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/10/2015] [Indexed: 12/16/2022]
Abstract
Diseases in articular cartilages affect millions of people. Despite the relatively simple biochemical and cellular composition of articular cartilages, the self-repair ability of cartilage is limited. Successful cartilage tissue engineering requires intricately coordinated interactions between matrerials, cells, biological factors, and phycial/mechanical factors, and still faces a multitude of challenges. This article presents an overview of the cartilage biology, current treatments, recent advances in the materials, biological factors, and cells used in cartilage tissue engineering/regeneration, with strong emphasis on the perspectives of gene regulation (e.g., microRNA) and gene therapy.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| | - Yu-Chen Hu
- Department of Chemical Engineering; National Tsing Hua University; Hsinchu Taiwan 300
| |
Collapse
|
215
|
Abina SHB, Gaspar HB, Blondeau J, Caccavelli L, Charrier S, Buckland K, Picard C, Six E, Himoudi N, Gilmour K, McNicol AM, Hara H, Xu-Bayford J, Rivat C, Touzot F, Mavilio F, Lim A, Treluyer JM, Héritier S, Lefrere F, Magalon J, Pengue-Koyi I, Honnet G, Blanche S, Sherman EA, Male F, Berry C, Malani N, Bushman FD, Fischer A, Thrasher AJ, Galy A, Cavazzana M. Outcomes following gene therapy in patients with severe Wiskott-Aldrich syndrome. JAMA 2015; 313:1550-63. [PMID: 25898053 PMCID: PMC4942841 DOI: 10.1001/jama.2015.3253] [Citation(s) in RCA: 283] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IMPORTANCE Wiskott-Aldrich syndrome is a rare primary immunodeficiency associated with severe microthrombocytopenia. Partially HLA antigen-matched allogeneic hematopoietic stem cell (HSC) transplantation is often curative but is associated with significant comorbidity. OBJECTIVE To assess the outcomes and safety of autologous HSC gene therapy in Wiskott-Aldrich syndrome. DESIGN, SETTING, AND PARTICIPANTS Gene-corrected autologous HSCs were infused in 7 consecutive patients with severe Wiskott-Aldrich syndrome lacking HLA antigen-matched related or unrelated HSC donors (age range, 0.8-15.5 years; mean, 7 years) following myeloablative conditioning. Patients were enrolled in France and England and treated between December 2010 and January 2014. Follow-up of patients in this intermediate analysis ranged from 9 to 42 months. INTERVENTION A single infusion of gene-modified CD34+ cells with an advanced lentiviral vector. MAIN OUTCOMES AND MEASURES Primary outcomes were improvement at 24 months in eczema, frequency and severity of infections, bleeding tendency, and autoimmunity and reduction in disease-related days of hospitalization. Secondary outcomes were improvement in immunological and hematological characteristics and evidence of safety through vector integration analysis. RESULTS Six of the 7 patients were alive at the time of last follow-up (mean and median follow-up, 28 months and 27 months, respectively) and showed sustained clinical benefit. One patient died 7 months after treatment of preexisting drug-resistant herpes virus infection. Eczema and susceptibility to infections resolved in all 6 patients. Autoimmunity improved in 5 of 5 patients. No severe bleeding episodes were recorded after treatment, and at last follow-up, all 6 surviving patients were free of blood product support and thrombopoietic agonists. Hospitalization days were reduced from a median of 25 days during the 2 years before treatment to a median of 0 days during the 2 years after treatment. All 6 surviving patients exhibited high-level, stable engraftment of functionally corrected lymphoid cells. The degree of myeloid cell engraftment and of platelet reconstitution correlated with the dose of gene-corrected cells administered. No evidence of vector-related toxicity was observed clinically or by molecular analysis. CONCLUSIONS AND RELEVANCE This study demonstrated the feasibility of the use of gene therapy in patients with Wiskott-Aldrich syndrome. Controlled trials with larger numbers of patients are necessary to assess long-term outcomes and safety.
Collapse
Affiliation(s)
- Salima Hacein-Bey Abina
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
- UTCBS CNRS 8258- INSERM U1022, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
- Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, AP-HP, 78, rue du Général-Leclerc, 94270 Le-Kremlin-Bicêtre, France
| | - H. Bobby Gaspar
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Johanna Blondeau
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Laure Caccavelli
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Sabine Charrier
- INSERM, U951; University of Evry, UMR_S951; Molecular Immunology and Innovative Biotherapies, Genethon, Evry, F-91002 France
- Genethon, Evry, F-91002 France
| | - Karen Buckland
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Capucine Picard
- Centre d’Étude des Déficits Immunitaires, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Emmanuelle Six
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- INSERM UMR 1163, Laboratory of human lymphohematopoiesis, Paris, France
| | - Nourredine Himoudi
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Kimberly Gilmour
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Anne-Marie McNicol
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Havinder Hara
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Jinhua Xu-Bayford
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Christine Rivat
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Fabien Touzot
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Annick Lim
- Groupe Immunoscope, Immunology Department, Institut Pasteur, Paris, France
| | - Jean-Marc Treluyer
- Clinical research Center Necker-Enfants Malades and Cochin Hospital Assistance Publique, Hôpitaux de Paris, Paris Descartes University
| | - Sébastien Héritier
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francois Lefrere
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jeremy Magalon
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Isabelle Pengue-Koyi
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | | | - Stéphane Blanche
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Eric A. Sherman
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Frances Male
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Charles Berry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Nirav Malani
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Frederic D. Bushman
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Alain Fischer
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM UMR 1163, Laboratory of human lymphohematopoiesis, Paris, France
- Collège de France, Paris, France
| | - Adrian J. Thrasher
- Section of Molecular and Cellular Immunology, University College London Institute of Child Health, London, UK
- Dept of Clinical Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Anne Galy
- INSERM, U951; University of Evry, UMR_S951; Molecular Immunology and Innovative Biotherapies, Genethon, Evry, F-91002 France
- Genethon, Evry, F-91002 France
| | - Marina Cavazzana
- Biotherapy Department, Necker Children’s Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
- Paris Descartes – Sorbonne Paris Cité University, Imagine Institute, Paris, France
- Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM UMR 1163, Laboratory of human lymphohematopoiesis, Paris, France
- To whom correspondence should be addressed: Marina Cavazzana, MD, PhD: Address: Biotherapy Department, Necker Children’s Hospital, 149 rue de Sèvres, 75015 Paris, France. Phone number: 00.33(1)44.49.50.68,
| |
Collapse
|
216
|
Multhaup MM, Podetz-Pedersen KM, Karlen AD, Olson ER, Gunther R, Somia NV, Blazar BR, Cowan MJ, McIvor RS. Role of transgene regulation in ex vivo lentiviral correction of artemis deficiency. Hum Gene Ther 2015; 26:232-43. [PMID: 25738323 DOI: 10.1089/hum.2014.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Artemis is a single-stranded endonuclease, deficiency of which results in a radiation-sensitive form of severe combined immunodeficiency (SCID-A) most effectively treated by allogeneic hematopoietic stem cell (HSC) transplantation and potentially treatable by administration of genetically corrected autologous HSCs. We previously reported cytotoxicity associated with Artemis overexpression and subsequently characterized the human Artemis promoter with the intention to provide Artemis expression that is nontoxic yet sufficient to support immunodevelopment. Here we compare the human Artemis promoter (APro) with the moderate-strength human phosphoglycerate kinase (PGK) promoter and the strong human elongation factor-1α (EF1α) promoter to regulate expression of Artemis after ex vivo lentiviral transduction of HSCs in a murine model of SCID-A. Recipient animals treated with the PGK-Artemis vector exhibited moderate repopulation of their immune compartment, yet demonstrated a defective proliferative T lymphocyte response to in vitro antigen stimulation. Animals treated with the EF1α-Artemis vector displayed high levels of T lymphocytes but an absence of B lymphocytes and deficient lymphocyte function. In contrast, ex vivo transduction with the APro-Artemis vector supported effective immune reconstitution to wild-type levels, resulting in fully functional T and B lymphocyte responses. These results demonstrate the importance of regulated Artemis expression in immune reconstitution of Artemis-deficient SCID.
Collapse
Affiliation(s)
- Megan M Multhaup
- 1 Department of Genetics, Cell Biology, and Development, University of Minnesota , Minneapolis, MN 55455
| | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Faster T-cell development following gene therapy compared with haploidentical HSCT in the treatment of SCID-X1. Blood 2015; 125:3563-9. [PMID: 25869287 DOI: 10.1182/blood-2014-12-616003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/01/2015] [Indexed: 12/22/2022] Open
Abstract
During the last decade, gene therapy via ex vivo gene transfer into autologous hematopoietic stem cells has emerged as a convincing therapy for severe combined immunodeficiency caused by ILR2G mutation (SCID-X1) despite the occurrence of genotoxicity caused by the integration of first-generation retroviral vectors. However, the place of gene therapy among the therapeutic armamentarium remains to be defined. We retrospectively analyze and compare clinical outcomes and immune reconstitution in 13 consecutive SCID-X1 patients having undergone haploidentical hematopoietic stem cell transplantation (HSCT) and 14 SCID-X1 patients treated with gene therapy over the same period at a single center level: the Necker Children's Hospital (Paris, France). Our results show a clear advantage in terms of T-cell development of gene therapy over HSCT with a mismatched donor. Patients treated with gene therapy display a faster T-cell reconstitution and a better long-term thymic output. Interestingly, this advantage of gene therapy vs haploidentical HSCT seems to be independent of the existence of clinical graft-versus-host disease in the latter condition. If data of safety are confirmed over the long term, gene therapy for SCID-X1 appears to be an equal, if not superior, alternative to haploidentical HSCT.
Collapse
|
218
|
Adeno-associated virus vector-based gene therapy for monogenetic metabolic diseases of the liver. J Pediatr Gastroenterol Nutr 2015; 60:433-40. [PMID: 25594875 DOI: 10.1097/mpg.0000000000000703] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Liver-based metabolic diseases account for a substantial burden of childhood diseases. In most patients, treatment is often limited to supportive measures and liver transplantation is ultimately required. Even despite the excellent long-term outcome of liver transplantation, the procedure is associated with a significant morbidity and mortality. Gene therapy, in contrast, has great potential to save lives, improve the quality of life, and offer few risks and adverse effects compared with present therapies including liver transplantation. The most promising results to date in liver gene transfer have been achieved with adeno-associated virus. Although safety issues, such as immunogenicity of vector and/or transgene product, remain an important concern, gene therapy is ready for clinical trials in adults and adolescents. Developing and testing safe approaches for efficient and long-term stable applications in newborns and small children, such as targeted integration and gene correction, is one of the remaining future challenges.
Collapse
|
219
|
Fischer A, Hacein-Bey Abina S, Touzot F, Cavazzana M. Gene therapy for primary immunodeficiencies. Clin Genet 2015; 88:507-15. [PMID: 25708106 DOI: 10.1111/cge.12576] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/09/2015] [Accepted: 02/16/2015] [Indexed: 12/15/2022]
Abstract
Gene therapy has effectively entered Medicine via the field of primary immunodeficiencies (PID). Because hematopoietic stem cells are accessible and because it was understood that genetic correction of lymphocyte progenitor cells carrying a genetic defect impairing differentiation, could result in the production of long-lived T lymphocytes, it was reasoned that ex vivo gene transfer in hematopoietic cells could lead to disease phenotype correction. Retroviral vectors were designed to ex vivo transduce such cells. This has indeed been shown to lead to sustained correction of the T cell immunodeficiency associated with two forms of severe combined immunodeficiencies (SCID) for now more than ten years. Occurrence in some patients of genotoxicity related to retroviral vectors integration close to and transactivation of oncogenes has led to the development of retroviral vectors devoid of its enhancer element. Results of recent trials performed for several forms of PID indeed suggest that their use is both safe and efficacious. It is thus anticipated that their application to the treatment of many more life threatening PID will be developed over the coming years.
Collapse
Affiliation(s)
- A Fischer
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France.,INSERM UMR 1163, Institut Imagine, Paris, France.,Collège de France, Paris, France
| | - S Hacein-Bey Abina
- UTCBS CNRS 8258 - INSERM U1022, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France.,Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Le-Kremlin-Bicêtre, France
| | - F Touzot
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,INSERM UMR 1163, Institut Imagine, Paris, France.,Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - M Cavazzana
- Paris Descartes - Sorbonne Paris Cité University, Imagine Institute, Paris, France.,INSERM UMR 1163, Institut Imagine, Paris, France.,Biotherapy Department, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| |
Collapse
|
220
|
Hu Y, O'Boyle K, Palmer D, Ng P, Sutton RE. High-level production of replication-defective human immunodeficiency type 1 virus vector particles using helper-dependent adenovirus vectors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15004. [PMID: 26029715 PMCID: PMC4444993 DOI: 10.1038/mtm.2015.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 11/09/2022]
Abstract
Gene transfer vectors based upon human immunodeficiency virus type 1 (HIV) are widely used in bench research applications and increasingly in clinical investigations, both to introduce novel genes but also to reduce expression of unwanted genes of the host and pathogen. At present, the vast majority of HIV-based vector supernatants are produced in 293T cells by cotransfection of up to five DNA plasmids, which is subject to variability and difficult to scale. Here we report the development of a HIV-based vector production system that utilizes helper-dependent adenovirus (HDAd). All necessary HIV vector components were inserted into one or more HDAds, which were then amplified to very high titers of ~1013 vp/ml. These were then used to transduce 293-based cells to produce HIV-based vector supernatants, and resultant VSV G-pseudotyped lentiviral vector (LV) titers and total IU were 10- to 30-fold higher, compared to plasmid transfection. Optimization of HIV-based vector production depended upon maximizing expression of all HIV vector components from HDAd. Supernatants contained trace amounts of HDAd but were free of replication-competent lentivirus. This production method should be applicable to other retroviral vector systems. Scalable production of HIV-based vectors using this two-step procedure should facilitate their clinical advancement.
Collapse
Affiliation(s)
- Yani Hu
- Division of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut, USA
| | - Kaitlin O'Boyle
- Division of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut, USA
| | - Donna Palmer
- Department of Molecular and Human Genetics, Baylor College of Medicine , Houston, Texas, USA
| | - Philip Ng
- Department of Molecular and Human Genetics, Baylor College of Medicine , Houston, Texas, USA
| | - Richard E Sutton
- Division of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut, USA
| |
Collapse
|
221
|
Calero-Garcia M, Gaspar HB. Gene Therapy for SCID. CURRENT PEDIATRICS REPORTS 2015. [DOI: 10.1007/s40124-014-0069-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
222
|
Barbero N, Magistris C, Quagliotto P, Bonandini L, Barolo C, Buscaino R, Compari C, Contardi L, Fisicaro E, Viscardi G. Synthesis, Physicochemical Characterization, and Interaction with DNA of Long-Alkyl-Chain Gemini Pyridinium Surfactants. Chempluschem 2015; 80:952-962. [PMID: 31973260 DOI: 10.1002/cplu.201500007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Indexed: 11/10/2022]
Abstract
Pyridinium gemini surfactants with hexadecyl chains linked to nitrogen atoms and a tuned aliphatic spacer that bridges the two pyridinium polar heads in 2,2'-positions have been synthesized and characterized. A multitechnique approach allowed us to study the aggregation behavior, using conductivity, surface tension, and fluorescence. Graphs of the specific conductivity (κ) versus the surfactant molar concentration (C), and graphs of the molar conductivity (Λ) versus C0.5 suggest pre-aggregation phenomena of these amphiphiles at very low concentration. The trends of Amin as a function of the spacer length confirm the hypothesis of a conformational change of the molecule with four methylene groups as spacer owing to stacking interactions between the two pyridinium rings mediated by the counterion. Moreover, the trends of Amin and counterion binding (β) suggest that the spacer must be longer than eight carbon atoms to fold efficiently toward the micellar core. The opportunity to tune the surfactant structure and aggregation properties make those surfactants-particularly the long-chain ones for which the DNA complexing ability was shown by means of atomic force microscopy (AFM) imaging-desirable candidates for gene-delivery experiments.
Collapse
Affiliation(s)
- Nadia Barbero
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Claudio Magistris
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Pierluigi Quagliotto
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Luca Bonandini
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Claudia Barolo
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Roberto Buscaino
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| | - Carlotta Compari
- Dipartimento di Farmacia-Università di Parma, Parco Area delle Scienze 27A, 43124 Parma (Italy)
| | - Laura Contardi
- Dipartimento di Farmacia-Università di Parma, Parco Area delle Scienze 27A, 43124 Parma (Italy)
| | - Emilia Fisicaro
- Dipartimento di Farmacia-Università di Parma, Parco Area delle Scienze 27A, 43124 Parma (Italy)
| | - Guido Viscardi
- Dipartimento di Chimica, Interdepartmental "Nanostructured Surfaces and Interfaces", NIS Centre-Università di Torino, Via Pietro Giuria 7, 10125 Torino (Italy)
| |
Collapse
|
223
|
Felsburg PJ, De Ravin SS, Malech HL, Sorrentino BP, Burtner C, Kiem HP. Gene therapy studies in a canine model of X-linked severe combined immunodeficiency. HUM GENE THER CL DEV 2015; 26:50-6. [PMID: 25603151 DOI: 10.1089/humc.2015.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Since the occurrence of T cell leukemias in the original human γ-retroviral gene therapy trials for X-linked severe combined immunodeficiency (XSCID), considerable effort has been devoted to developing safer vectors. This review summarizes gene therapy studies performed in a canine model of XSCID to evaluate the efficacy of γ-retroviral, lentiviral, and foamy viral vectors for treating XSCID and a novel method of vector delivery. These studies demonstrate that durable T cell reconstitution and thymopoiesis with no evidence of any serious adverse events and, in contrast to the human XSCID patients, sustained marking in myeloid cells and B cells with reconstitution of normal humoral immune function can be achieved for up to 5 years without any pretreatment conditioning. The presence of sustained levels of gene-marked T cells, B cells, and more importantly myeloid cells for almost 5 years is highly suggestive of transduction of either multipotent hematopoietic stem cells or very primitive committed progenitors.
Collapse
Affiliation(s)
- Peter J Felsburg
- 1 Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, PA 19104
| | | | | | | | | | | |
Collapse
|
224
|
Abstract
Gene therapy has a growing research potential particularly in the field of ophthalmic and retinal diseases owing to three main characteristics of the eye; accessibility in terms of injections and surgical interventions, its immune-privileged status facilitating the accommodation to the antigenicity of a viral vector, and tight blood-ocular barriers which save other organs from unwanted contamination. Gene therapy has tremendous potential for different ocular diseases. In fact, the perspective of gene therapy in the field of eye research does not confine to exclusive monogenic ophthalmic problems and it has the potential to include gene based pharmacotherapies for non-monogenic problems such as age related macular disease and diabetic retinopathy. The present article has focused on how gene transfer into the eye has been developed and used to treat retinal disorders with no available therapy at present.
Collapse
|
225
|
Preparation and Testing of Quaternized Chitosan Nanoparticles as Gene Delivery Vehicles. Appl Biochem Biotechnol 2015; 175:3244-57. [DOI: 10.1007/s12010-015-1483-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 01/05/2015] [Indexed: 02/04/2023]
|
226
|
O'Reilly M, Federoff HJ, Fong Y, Kohn DB, Patterson AP, Ahmed N, Asokan A, Boye SE, Crystal RG, De Oliveira S, Gargiulo L, Harper SQ, Ikeda Y, Jambou R, Montgomery M, Prograis L, Rosenthal E, Sterman DH, Vandenberghe LH, Zoloth L, Abedi M, Adair J, Adusumilli PS, Goins WF, Gray J, Monahan P, Popplewell L, Sena-Esteves M, Tannous B, Weber T, Wierda W, Gopal-Srivastava R, McDonald CL, Rosenblum D, Corrigan-Curay J. Gene therapy: charting a future course--summary of a National Institutes of Health Workshop, April 12, 2013. Hum Gene Ther 2015; 25:488-97. [PMID: 24773122 DOI: 10.1089/hum.2014.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recently, the gene therapy field has begun to experience clinical successes in a number of different diseases using various approaches and vectors. The workshop Gene Therapy: Charting a Future Course, sponsored by the National Institutes of Health (NIH) Office of Biotechnology Activities, brought together early and mid-career researchers to discuss the key scientific challenges and opportunities, ethical and communication issues, and NIH and foundation resources available to facilitate further clinical advances.
Collapse
Affiliation(s)
- Marina O'Reilly
- 1 Office of Science Policy, Office of the Director, National Institutes of Health , Bethesda, MD 20892
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Abstract
Fifty years ago, in 1964, our understanding of the immune system was very rudimentary. Gell and Coombs had just described classes of hypersensitivity reactions, and Bruton had described and commenced immunoglobulin replacement in agammaglobulinaemia. The distinction between T and B cells was not identified and characterised until the 1960s and 1970s. This was followed by increasing recognition of T and B cell collaboration in immune responses and identification of significant immunodeficiencies. CD4 and CD8 T cells were only recognised in the 1970s and 1980s. We now know of five CD4 subsets; dysfunction of each is associated with different disorders. By 2014, advances in technology have enabled identification of the genetic basis of over 240 primary immunodeficiencies. Research into the gut microbiome and vitamin D holds promise for the understanding, treatment and prevention of autoimmune and allergic diseases. Immunoglobulin preparations for the treatment of antibody deficiencies improved with the development of preparations for intravenous then subcutaneous administration, giving patients choice and the ability for home-based treatment, especially if experiencing infusion associated adverse effects. Newborn screening for severe combined immunodeficiency is a reality. Improvements in haemopoietic stem cell transplantation and now gene therapy, albeit still only available in the research setting, are improving long-term survival in primary immunodeficiencies. Biologic therapeutic agents are improving the control of autoimmune disease but potentially leading to secondary immunodeficiency, increasing the risk of opportunistic infection and malignancy. It is an exciting time.
Collapse
Affiliation(s)
- Melanie Wong
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
228
|
Hladun R, Badell I, González M, Martínez A, Sánchez de Toledo J, Olivé M, González M, Elorza I, Díaz de Heredia C. Survival analysis of hematopoietic stem cell transplantation in children with primary immunodeficiency in Spain. ANALES DE PEDIATRÍA (ENGLISH EDITION) 2015. [DOI: 10.1016/j.anpede.2014.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
229
|
Abstract
The ability to remove blood cells, including hematopoietic stem cells (HSCs), from a person and then re-transplant them (hematopoietic stem cell transplantation (HSCT) is a well-established treatment paradigm that can be used in both the autologous setting or in the allogeneic setting. Using allogeneic HSCT can cure different genetic diseases of the blood but has significant limitations. An alternative to allogeneic HSCT is to transplant genetically modified HSCs instead. A powerful approach to the precision modification of HSCs is to use genome editing whereby the genome is modified with spatial precision (at an exact location) in the genome and sometimes with nucleotide precision (the exact nucleotide changes are introduced). The progress and challenges of genome editing of blood are discussed.
Collapse
Affiliation(s)
- Matthew H Porteus
- Dept. of Pediatrics MC5462, Stanford University, Stanford, CA 94305,
| |
Collapse
|
230
|
Liu M, Maurano MT, Wang H, Qi H, Song CZ, Navas PA, Emery DW, Stamatoyannopoulos JA, Stamatoyannopoulos G. Genomic discovery of potent chromatin insulators for human gene therapy. Nat Biotechnol 2015; 33:198-203. [PMID: 25580597 DOI: 10.1038/nbt.3062] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 10/09/2014] [Indexed: 12/29/2022]
Abstract
Insertional mutagenesis and genotoxicity, which usually manifest as hematopoietic malignancy, represent major barriers to realizing the promise of gene therapy. Although insulator sequences that block transcriptional enhancers could mitigate or eliminate these risks, so far no human insulators with high functional potency have been identified. Here we describe a genomic approach for the identification of compact sequence elements that function as insulators. These elements are highly occupied by the insulator protein CTCF, are DNase I hypersensitive and represent only a small minority of the CTCF recognition sequences in the human genome. We show that the elements identified acted as potent enhancer blockers and substantially decreased the risk of tumor formation in a cancer-prone animal model. The elements are small, can be efficiently accommodated by viral vectors and have no detrimental effects on viral titers. The insulators we describe here are expected to increase the safety of gene therapy for genetic diseases.
Collapse
Affiliation(s)
- Mingdong Liu
- Division of Medical Genetics, University of Washington, Seattle, Washington, USA
| | - Matthew T Maurano
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Hao Wang
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Heyuan Qi
- 1] Division of Medical Genetics, University of Washington, Seattle, Washington, USA. [2] Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Chao-Zhong Song
- 1] Division of Medical Genetics, University of Washington, Seattle, Washington, USA. [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Patrick A Navas
- Division of Medical Genetics, University of Washington, Seattle, Washington, USA
| | - David W Emery
- 1] Division of Medical Genetics, University of Washington, Seattle, Washington, USA. [2] Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - John A Stamatoyannopoulos
- 1] Department of Genome Sciences, University of Washington, Seattle, Washington, USA. [2] Department of Medicine, University of Washington, Seattle, Washington, USA
| | - George Stamatoyannopoulos
- 1] Division of Medical Genetics, University of Washington, Seattle, Washington, USA. [2] Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
231
|
Abstract
Epstein-Barr virus (EBV) is associated with a range of malignancies involving B cells, T cells, natural killer (NK) cells, epithelial cells, and smooth muscle. All of these are associated with the latent life cycles of EBV, but the pattern of latency-associated viral antigens expressed in tumor cells depends on the type of tumor. EBV-specific T cells (EBVSTs) have been explored as prophylaxis and therapy for EBV-associated malignancies for more than two decades. EBVSTs have been most successful as prophylaxis and therapy for post-transplant lymphoproliferative disease (PTLD) , which expresses the full array of latent EBV antigens (type 3 latency), in hematopoietic stem-cell transplant (HSCT) recipients. While less effective, clinical studies have also demonstrated their therapeutic potential for PTLD post-solid organ transplant and for EBV-associated malignancies such as Hodgkin's lymphoma, non-Hodgkin's lymphoma, and nasopharyngeal carcinoma (NPC) that express a limited array of latent EBV antigens (type 2 latency). Several approaches are actively being pursued to improve the antitumor activity of EBVSTs including activation and expansion of T cells specific for the EBV antigens expressed in type 2 latency, genetic approaches to render EBVSTs resistant to the immunosuppressive tumor environment, and combination approaches with other immune-modulating modalities. Given the recent advances and renewed interest in cell therapy, we hope that EBVSTs will become an integral part of our treatment armamentarium against EBV-positive malignancies in the near-future.
Collapse
|
232
|
Mikkelsen JG. Nonviral Gene Therapy—The Challenge of Mobilizing DNA. SOMATIC GENOME MANIPULATION 2015:69-104. [DOI: 10.1007/978-1-4939-2389-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
233
|
Abstract
Lentiviral vectors are the workhorses of modern cell biology. They can infect a wide variety of cells including nondividing cells and stem cells. They integrate into the genome of infected cells leading to stable expression. It is easy to transduce 100 % of the cells in a culture and possible to infect cells simultaneously with multiple vectors, greatly facilitating studies on malignant transformation. We present simple protocols to produce and titrate lentiviral vectors, infect mammary epithelial cells, and check for contamination with replication-competent viruses.
Collapse
|
234
|
Perforin gene transfer into hematopoietic stem cells improves immune dysregulation in murine models of perforin deficiency. Mol Ther 2014; 23:737-45. [PMID: 25523759 PMCID: PMC4395774 DOI: 10.1038/mt.2014.242] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/06/2014] [Indexed: 12/11/2022] Open
Abstract
Defects in perforin lead to the failure of T and NK cell cytotoxicity, hypercytokinemia, and the immune dysregulatory condition known as familial hemophagocytic lymphohistiocytosis (FHL). The only curative treatment is allogeneic hematopoietic stem cell transplantation which carries substantial risks. We used lentiviral vectors (LV) expressing the human perforin gene, under the transcriptional control of the ubiquitous phosphoglycerate kinase promoter or a lineage-specific perforin promoter, to correct the defect in different murine models. Following LV-mediated gene transfer into progenitor cells from perforin-deficient mice, we observed perforin expression in mature T and NK cells, and there was no evidence of progenitor cell toxicity when transplanted into irradiated recipients. The resulting perforin-reconstituted NK cells showed partial recovery of cytotoxicity, and we observed full recovery of cytotoxicity in polyclonal CD8+ T cells. Furthermore, reconstituted T cells with defined antigen specificity displayed normal cytotoxic function against peptide-loaded targets. Reconstituted CD8+ lymphoblasts had reduced interferon-γ secretion following stimulation in vitro, suggesting restoration of normal immune regulation. Finally, upon viral challenge, mice with >30% engraftment of gene-modified cells exhibited reduction of cytokine hypersecretion and cytopenias. This study demonstrates the potential of hematopoietic stem cell gene therapy as a curative treatment for perforin-deficient FHL.
Collapse
|
235
|
Ebadi M, Aghamohammadi A, Rezaei N. Primary immunodeficiencies: a decade of shifting paradigms, the current status and the emergence of cutting-edge therapies and diagnostics. Expert Rev Clin Immunol 2014; 11:117-39. [DOI: 10.1586/1744666x.2015.995096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
236
|
Gene therapy for inherited muscle diseases: where genetics meets rehabilitation medicine. Am J Phys Med Rehabil 2014; 93:S97-107. [PMID: 25313664 DOI: 10.1097/phm.0000000000000138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of clinical vectors to correct genetic mutations that cause inherited myopathies and related disorders of skeletal muscle is advancing at an impressive rate. Adeno-associated virus vectors are attractive for clinical use because (1) adeno-associated viruses do not cause human disease and (2) these vectors are able to persist for years. New vectors are now becoming available as gene therapy delivery tools, and recent preclinical experiments have demonstrated the feasibility, safety, and efficacy of gene therapy with adeno-associated virus for long-term correction of muscle pathology and weakness in myotubularin-deficient canine and murine disease models. In this review, recent advances in the application of gene therapies to treat inherited muscle disorders are presented, including Duchenne muscular dystrophy and x-linked myotubular myopathy. Potential areas for therapeutic synergies between rehabilitation medicine and genetics are also discussed.
Collapse
|
237
|
Kim S, Kim N, Presson AP, Metzger ME, Bonifacino AC, Sehl M, Chow SA, Crooks GM, Dunbar CE, An DS, Donahue RE, Chen ISY. Dynamics of HSPC repopulation in nonhuman primates revealed by a decade-long clonal-tracking study. Cell Stem Cell 2014; 14:473-85. [PMID: 24702996 DOI: 10.1016/j.stem.2013.12.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/13/2013] [Accepted: 12/18/2013] [Indexed: 02/07/2023]
Abstract
In mice, clonal tracking of hematopoietic stem cells (HSCs) has revealed variations in repopulation characteristics. However, it is unclear whether similar properties apply in primates. Here, we examined this issue through tracking of thousands of hematopoietic stem and progenitor cells (HSPCs) in rhesus macaques for up to 12 years. Approximately half of the clones analyzed contributed to long-term repopulation (over 3-10 years), arising in sequential groups and likely representing self-renewing HSCs. The remainder contributed primarily for the first year. The long-lived clones could be further subdivided into functional groups contributing primarily to myeloid, lymphoid, or both myeloid and lymphoid lineages. Over time, the 4%-10% of clones with robust dual lineage contribution predominated in repopulation. HSPCs expressing a CCR5 shRNA transgene behaved similarly to controls. Our study therefore documents HSPC behavior in a clinically relevant model over a long time frame and provides a substantial system-level data set that is a reference point for future work.
Collapse
Affiliation(s)
- Sanggu Kim
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Namshin Kim
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, 111 Gwahangno, Yuseong-gu, Daejeon 305-806, Korea
| | - Angela P Presson
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mark E Metzger
- Hematology Branch, National Heart, Lung and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Aylin C Bonifacino
- Hematology Branch, National Heart, Lung and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Mary Sehl
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biomathematics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samson A Chow
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gay M Crooks
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cynthia E Dunbar
- Hematology Branch, National Heart, Lung and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Dong Sung An
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Nursing, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Robert E Donahue
- Hematology Branch, National Heart, Lung and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Irvin S Y Chen
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
238
|
Alpharetroviral vectors: from a cancer-causing agent to a useful tool for human gene therapy. Viruses 2014; 6:4811-38. [PMID: 25490763 PMCID: PMC4276931 DOI: 10.3390/v6124811] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/07/2014] [Accepted: 11/20/2014] [Indexed: 12/24/2022] Open
Abstract
Gene therapy using integrating retroviral vectors has proven its effectiveness in several clinical trials for the treatment of inherited diseases and cancer. However, vector-mediated adverse events related to insertional mutagenesis were also observed, emphasizing the need for safer therapeutic vectors. Paradoxically, alpharetroviruses, originally discovered as cancer-causing agents, have a more random and potentially safer integration pattern compared to gammaretro- and lentiviruses. In this review, we provide a short overview of the history of alpharetroviruses and explain how they can be converted into state-of-the-art gene delivery tools with improved safety features. We discuss development of alpharetroviral vectors in compliance with regulatory requirements for clinical translation, and provide an outlook on possible future gene therapy applications. Taken together, this review is a broad overview of alpharetroviral vectors spanning the bridge from their parental virus discovery to their potential applicability in clinical settings.
Collapse
|
239
|
Kohn DB. Eliminating SCID row: new approaches to SCID. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:475-480. [PMID: 25696897 DOI: 10.1182/asheducation-2014.1.475] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Treatments for patients with SCID by hematopoietic stem cell transplantation (HSCT) have changed this otherwise lethal primary immune deficiency disorder into one with an increasingly good prognosis. SCID has been the paradigm disorder supporting many key advances in the field of HSCT, with first-in-human successes with matched sibling, haploidentical, and matched unrelated donor allogeneic transplantations. Nevertheless, the optimal approaches for HSCT are still being defined, including determining the optimal stem cell sources, the use and types of pretransplantation conditioning, and applications for SCID subtypes associated with radiosensitivity, for patients with active viral infections and for neonates. Alternatively, autologous transplantation after ex vivo gene correction (gene therapy) has been applied successfully to the treatment of adenosine deaminase-deficient SCID and X-linked SCID by vector-mediated gene addition. Gene therapy holds the prospect of avoiding risks of GVHD and would allow each patient to be their own donor. New approaches to gene therapy by gene correction in autologous HSCs using site-specific endonuclease-mediated homology-driven gene repair are under development. With newborn screening becoming more widely adopted to detect SCID patients before they develop complications, the prognosis for SCID is expected to improve further. This chapter reviews recent advances and ongoing controversies in allogeneic and autologous HSCT for SCID.
Collapse
Affiliation(s)
- Donald B Kohn
- Departments of Microbiology, Immunology, and Molecular Genetics and Pediatrics; David Geffen School of Medicine, Mattel Children's Hospital; and Eli & Edythe Broad Center for Regenerative Medicine and Stem Cells, University of California, Los Angeles, CA
| |
Collapse
|
240
|
|
241
|
Avci-Adali M, Behring A, Steinle H, Keller T, Krajeweski S, Schlensak C, Wendel HP. In vitro synthesis of modified mRNA for induction of protein expression in human cells. J Vis Exp 2014:e51943. [PMID: 25489992 DOI: 10.3791/51943] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The exogenous delivery of coding synthetic messenger RNA (mRNA) for induction of protein synthesis in desired cells has enormous potential in the fields of regenerative medicine, basic cell biology, treatment of diseases, and reprogramming of cells. Here, we describe a step by step protocol for generation of modified mRNA with reduced immune activation potential and increased stability, quality control of produced mRNA, transfection of cells with mRNA and verification of the induced protein expression by flow cytometry. Up to 3 days after a single transfection with eGFP mRNA, the transfected HEK293 cells produce eGFP. In this video article, the synthesis of eGFP mRNA is described as an example. However, the procedure can be applied for production of other desired mRNA. Using the synthetic modified mRNA, cells can be induced to transiently express the desired proteins, which they normally would not express.
Collapse
Affiliation(s)
- Meltem Avci-Adali
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen;
| | - Andreas Behring
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| | - Heidrun Steinle
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| | - Timea Keller
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| | - Stefanie Krajeweski
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| | - Christian Schlensak
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| | - Hans P Wendel
- Department of Thoracic, Cardiac, and Vascular Surgery, University Hospital Tuebingen
| |
Collapse
|
242
|
Miyazaki S, Yamamoto H, Miyoshi N, Wu X, Ogawa H, Uemura M, Nishimura J, Hata T, Takemasa I, Mizushima T, Konno M, Doki Y, Mori M, Ishii H. A Cancer Reprogramming Method Using MicroRNAs as a Novel Therapeutic Approach against Colon Cancer: Research for Reprogramming of Cancer Cells by MicroRNAs. Ann Surg Oncol 2014; 22 Suppl 3:S1394-401. [PMID: 25384704 DOI: 10.1245/s10434-014-4217-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND We previously generated induced pluripotent stem cells by reprograming adipose stem cells through the introduction of microRNAs targeting four transcription factors (Oct3/4, Sox2, c-Myc, and Klf4). In this study, we aimed to reprogram cancer cells using microRNAs to explore their therapeutic potential. METHODS Mature microRNAs (mir-302a-d, 369-3p and 5p, and mir-200c, as needed) were introduced into colon cancer cells (DLD-1, RKO, and HCT116) using lipofection. RESULTS The transfected cells exhibited an embryonic stem cell-like morphology and expressed the undifferentiated marker genes Nanog, Oct3/4, SOX2, and Klf4, as well as tumor-related antigen-1-60. These cells expressed neurogenic or adipogenic markers, indicating that reprogramming of the cancer cells was partially successful. Moreover, we found that miRNA-expressing DLD-1 cells showed low proliferative activity in vitro and in vivo, accompanied by increased expression of the tumor suppressor genes p16 (ink4a) and p21 (waf1) . miRNA-expressing DLD-1 cells also exhibited enhanced sensitivity to 5-fluorouracil, possibly through the downregulation of multidrug-resistant protein 8. The reprogrammed cells from DLD-1, RKO, and HCT116 cells exhibited reduced c-Myc expression, in contrast to the high c-Myc expression in the induced pluripotent cancer cells that were generated using four transcription factors. CONCLUSIONS Our cancer reprogramming method employing simple lipofection of mature microRNAs is safe and well suited for clinical application, because it avoids integration of exogenous genes into the host genome and allows escape from augmentation of c-Myc gene expression.
Collapse
Affiliation(s)
- Susumu Miyazaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Norikatsu Miyoshi
- Department of Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Xin Wu
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hisataka Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Taishi Hata
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ichiro Takemasa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masamitsu Konno
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hideshi Ishii
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Cancer Profiling Discovery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
243
|
Braun CJ, Boztug K, Paruzynski A, Witzel M, Schwarzer A, Rothe M, Modlich U, Beier R, Göhring G, Steinemann D, Fronza R, Ball CR, Haemmerle R, Naundorf S, Kühlcke K, Rose M, Fraser C, Mathias L, Ferrari R, Abboud MR, Al-Herz W, Kondratenko I, Maródi L, Glimm H, Schlegelberger B, Schambach A, Albert MH, Schmidt M, von Kalle C, Klein C. Gene therapy for Wiskott-Aldrich syndrome--long-term efficacy and genotoxicity. Sci Transl Med 2014; 6:227ra33. [PMID: 24622513 DOI: 10.1126/scitranslmed.3007280] [Citation(s) in RCA: 415] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Wiskott-Aldrich syndrome (WAS) is characterized by microthrombocytopenia, immunodeficiency, autoimmunity, and susceptibility to malignancies. In our hematopoietic stem cell gene therapy (GT) trial using a γ-retroviral vector, 9 of 10 patients showed sustained engraftment and correction of WAS protein (WASP) expression in lymphoid and myeloid cells and platelets. GT resulted in partial or complete resolution of immunodeficiency, autoimmunity, and bleeding diathesis. Analysis of retroviral insertion sites revealed >140,000 unambiguous integration sites and a polyclonal pattern of hematopoiesis in all patients early after GT. Seven patients developed acute leukemia [one acute myeloid leukemia (AML), four T cell acute lymphoblastic leukemia (T-ALL), and two primary T-ALL with secondary AML associated with a dominant clone with vector integration at the LMO2 (six T-ALL), MDS1 (two AML), or MN1 (one AML) locus]. Cytogenetic analysis revealed additional genetic alterations such as chromosomal translocations. This study shows that hematopoietic stem cell GT for WAS is feasible and effective, but the use of γ-retroviral vectors is associated with a substantial risk of leukemogenesis.
Collapse
Affiliation(s)
- Christian Jörg Braun
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, 80337 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Niederer HA, Bangham CRM. Integration site and clonal expansion in human chronic retroviral infection and gene therapy. Viruses 2014; 6:4140-64. [PMID: 25365582 PMCID: PMC4246213 DOI: 10.3390/v6114140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/09/2014] [Accepted: 10/21/2014] [Indexed: 12/20/2022] Open
Abstract
Retroviral vectors have been successfully used therapeutically to restore expression of genes in a range of single-gene diseases, including several primary immunodeficiency disorders. Although clinical trials have shown remarkable results, there have also been a number of severe adverse events involving malignant outgrowth of a transformed clonal population. This clonal expansion is influenced by the integration site profile of the viral integrase, the transgene expressed, and the effect of the viral promoters on the neighbouring host genome. Infection with the pathogenic human retrovirus HTLV-1 also causes clonal expansion of cells containing an integrated HTLV-1 provirus. Although the majority of HTLV-1-infected people remain asymptomatic, up to 5% develop an aggressive T cell malignancy. In this review we discuss recent findings on the role of the genomic integration site in determining the clonality and the potential for malignant transformation of cells carrying integrated HTLV-1 or gene therapy vectors, and how these results have contributed to the understanding of HTLV-1 pathogenesis and to improvements in gene therapy vector safety.
Collapse
Affiliation(s)
- Heather A Niederer
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London W2 1PG, UK.
| | - Charles R M Bangham
- Department of Immunology, Wright-Fleming Institute, Imperial College London, London W2 1PG, UK.
| |
Collapse
|
245
|
Braun CJ, Witzel M, Paruzynski A, Boztug K, von Kalle C, Schmidt M, Klein C. Gene therapy for Wiskott-Aldrich Syndrome-Long-term reconstitution and clinical benefits, but increased risk for leukemogenesis. Rare Dis 2014; 2:e947749. [PMID: 26942098 PMCID: PMC4755244 DOI: 10.4161/21675511.2014.947749] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/06/2014] [Accepted: 07/16/2014] [Indexed: 01/14/2023] Open
Abstract
Wiskott-Aldrich-Syndrome (WAS) is a rare X-linked recessive disease caused by mutations of the WAS gene. It is characterized by immunodeficiency, autoimmunity, low numbers of small platelets (microthrombocytopenia) and a high risk of cancer, especially B cell lymphoma and leukemia.
Collapse
Affiliation(s)
- Christian Joerg Braun
- Dr. von Hauner Children's Hospital; Ludwig Maximilians University Munich ; Munich, Germany
| | - Maximilian Witzel
- Dr. von Hauner Children's Hospital; Ludwig Maximilians University Munich ; Munich, Germany
| | - Anna Paruzynski
- Department of Translational Oncology; National Center for Tumor Diseases and German Cancer Research Center; Heidelberg, Germany; New address: BioNTech AG; Mainz, Germany
| | - Kaan Boztug
- Department of Pediatric Hematology/Oncology; Hannover Medical School; Hannover, Germany; Present address: CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences/Department of Pediatrics and Adolescent Medicine, Medical University of Vienna; Vienna, Austria
| | - Christof von Kalle
- Department of Translational Oncology; National Center for Tumor Diseases and German Cancer Research Center ; Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology; National Center for Tumor Diseases and German Cancer Research Center ; Heidelberg, Germany
| | - Christoph Klein
- Dr. von Hauner Children's Hospital; Ludwig Maximilians University Munich ; Munich, Germany
| |
Collapse
|
246
|
Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a "safety switch" to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Front Pharmacol 2014; 5:235. [PMID: 25389405 PMCID: PMC4211380 DOI: 10.3389/fphar.2014.00235] [Citation(s) in RCA: 263] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 10/07/2014] [Indexed: 12/14/2022] Open
Abstract
Immune modulation has become a central element in many cancer treatments, and T cells genetically engineered to express chimeric antigen receptors (CAR) may provide a new approach to cancer immunotherapy. Autologous CAR T cells that have been re-directed toward tumor-associated antigens (TAA) have shown promising results in phase 1 clinical trials, with some patients undergoing complete tumor regression. However, this T-cell therapy must carefully balance effective T-cell activation, to ensure antitumor activity, with the potential for uncontrolled activation that may produce immunopathology. An inducible Caspase 9 (iCasp9) “safety switch” offers a solution that allows for the removal of inappropriately activated CAR T cells. The induction of iCasp9 depends on the administration of the small molecule dimerizer drug AP1903 and dimerization results in rapid induction of apoptosis in transduced cells, preferentially killing activated cells expressing high levels of transgene. The iCasp9 gene has been incorporated into vectors for use in preclinical studies and demonstrates effective and reliable suicide gene activity in phase 1 clinical trials. A third-generation CAR incorporating iCasp9 re-directs T cells toward the GD2 TAA. GD2 is over-expressed in melanoma and other malignancies of neural crest origin and the safety and activity of these GD2-iCAR T cells will be investigated in CARPETS and other actively recruiting phase 1 trials.
Collapse
Affiliation(s)
- Tessa Gargett
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia Adelaide, SA, Australia
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia Adelaide, SA, Australia ; Cancer Clinical Trials Unit, Royal Adelaide Hospital Adelaide, SA, Australia ; Discipline of Medicine, University of Adelaide Adelaide, SA, Australia
| |
Collapse
|
247
|
Hacein-Bey-Abina S, Pai SY, Gaspar HB, Armant M, Berry CC, Blanche S, Bleesing J, Blondeau J, de Boer H, Buckland KF, Caccavelli L, Cros G, De Oliveira S, Fernández KS, Guo D, Harris CE, Hopkins G, Lehmann LE, Lim A, London WB, van der Loo JCM, Malani N, Male F, Malik P, Marinovic MA, McNicol AM, Moshous D, Neven B, Oleastro M, Picard C, Ritz J, Rivat C, Schambach A, Shaw KL, Sherman EA, Silberstein LE, Six E, Touzot F, Tsytsykova A, Xu-Bayford J, Baum C, Bushman FD, Fischer A, Kohn DB, Filipovich AH, Notarangelo LD, Cavazzana M, Williams DA, Thrasher AJ. A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. N Engl J Med 2014; 371:1407-17. [PMID: 25295500 PMCID: PMC4274995 DOI: 10.1056/nejmoa1404588] [Citation(s) in RCA: 298] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).
Collapse
Affiliation(s)
- Salima Hacein-Bey-Abina
- From the Departments of Biotherapy (S.H.-B.-A., J. Blondeau, L.C., F.T., M.C.) and Immunology and Pediatric Hematology (S.B., G.C., D.M., B.N., C.P., F.T., A.F.) and the Centre d'Étude des Déficits Immunitaires (C.P.), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), the Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM (S.H.-B.-A., J. Blondeau, L.C., F.T., M.C.), Unité de Technologies Chimiques et Biologiques pour la Santé, Centre National de la Recherche Scientifique, 8258-INSERM Unité 1022, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes (S.H.-B.-A.), Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, AP-HP, Le Kremlin-Bicêtre (S.H.-B.-A.), Imagine Institute, Paris Descartes-Sorbonne Paris Cité University (S.B., J. Blondeau, L.C., D.M., B.N., C.P., E.S., A.F., M.C.), INSERM Unités Mixtes de Recherche 1163, Laboratory of Human Lymphohematopoiesis (J. Blondeau, L.C., E.S., F.T., A.F., M.C.), Groupe Immunoscope, Immunology Department, Institut Pasteur (A.L.), and Collège de France (A.F.) - all in Paris; Division of Hematology-Oncology (S.-Y.P., H.B., D.G., C.E.H., G.H., L.E.L., W.B.L., D.A.W.) and Division of Immunology (L.D.N.), Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute (S.-Y.P., D.G., L.E.L., W.B.L., D.A.W.), Harvard Medical School (S.-Y.P., M.A., L.E.L., W.B.L., J.R., L.E.S., A.T., L.D.N., D.A.W.), Center for Human Cell Therapy, Program in Cellular and Molecular Medicine, Boston Children's Hospital (M.A., J.R., L.E.S., A.T.), Division of Hematologic Malignancies, Dana-Farber Cancer Institute (J.R.), and the Manton Center for Orphan Disease Research (L.D.N.) - all in Boston; Great Ormond Street Hospital for Children NHS Foundation Trust (H.B.G., J.X.-B., A.J.T.) and Section of Molecular and Cellular Immunology, University College London Institute of Child Health (H.B.G., K.F.B., A
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Cyclosporin a and rapamycin relieve distinct lentiviral restriction blocks in hematopoietic stem and progenitor cells. Mol Ther 2014; 23:352-62. [PMID: 25270076 DOI: 10.1038/mt.2014.193] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/22/2014] [Indexed: 02/06/2023] Open
Abstract
Improving hematopoietic stem and progenitor cell (HSPC) permissiveness to HIV-derived lentiviral vectors (LVs) remains a challenge for the field of gene therapy as high vector doses and prolonged ex vivo culture are still required to achieve clinically relevant transduction levels. We report here that Cyclosporin A (CsA) and Rapamycin (Rapa) significantly improve LV gene transfer in human and murine HSPC. Both compounds increased LV but not gammaretroviral transduction and acted independently of calcineurin and autophagy. Improved gene transfer was achieved across all CD34(+) subpopulations, including in long-term SCID repopulating cells. Effects of CsA were specific of HSPC and opposite to its known impact on HIV replication. Mutating the Cyclophilin A binding pocket of the viral capsid (CA) further improved transduction in combination with CsA. Tracking of the LV genome fate revealed that CsA relieves a CA-dependent early block and increases integration, while Rapa acts early in LV infection independently of the viral CA. In agreement, only Rapa was able to improve transduction by an integrase-defective LV harboring wild-type CA. Overall, our findings pave the way for more efficient and sustainable LV gene therapy in human HSPCs and shed light on the multiple innate barriers specifically hampering LV transduction in these cells.
Collapse
|
249
|
Abou-El-Enein M, Bauer G, Reinke P, Renner M, Schneider CK. A roadmap toward clinical translation of genetically-modified stem cells for treatment of HIV. Trends Mol Med 2014; 20:632-42. [PMID: 25262540 DOI: 10.1016/j.molmed.2014.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 12/21/2022]
Abstract
During the past decade, successful gene therapies for immunodeficiencies were finally brought to the clinic. This was accomplished through new gene therapy vectors and improved procedures for genetic modification of autologous hematopoietic stem cells. For HIV, autologous hematopoietic stem cell (HSC) gene therapy with 'anti-HIV genes' promises a functional cure for the disease. However, to develop such a therapy and translate it into a clinical application is rather challenging. The risks and benefits of such a therapy have to be understood, and regulatory hurdles need to be overcome. In this joint paper by academic researchers and regulators, we are, therefore, outlining a high level roadmap for the early stage development of HSC gene therapy as a potential functional cure for HIV.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany.
| | - Gerhard Bauer
- University of California Davis, Institute For Regenerative Cures (IRC) Sacramento, CA, USA
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | - Christian K Schneider
- Formerly Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, UK; Danish Health and Medicines Authority, Axel Heides Gade 1, 2300 Copenhagen, Denmark; Twincore Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Straße 730625 Hannover, Germany
| |
Collapse
|
250
|
Wagemaker G. Lentiviral hematopoietic stem cell gene therapy in inherited metabolic disorders. Hum Gene Ther 2014; 25:862-5. [PMID: 25184354 DOI: 10.1089/hum.2014.102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
After more than 20 years of development, lentiviral hematopoietic stem cell gene therapy has entered the stage of initial clinical implementation for immune deficiencies and storage disorders. This brief review summarizes the development and applications, focusing on the lysosomal enzyme deficiencies, especially Pompe disease.
Collapse
Affiliation(s)
- Gerard Wagemaker
- Erasmus University Rotterdam, 3005 LA Rotterdam, The Netherlands
| |
Collapse
|