Gene therapy comes of age

Gene therapy: The power of persistence Nearly 50 years after the concept was first proposed, gene therapy is now considered a promising treatment option for several human diseases. The path to success has been long and tortuous. Serious adverse effects were encountered in early clinical studies, but this fueled basic research that led to safer and more efficient gene transfer vectors. Gene therapy in various forms has produced clinical benefits in patients with blindness, neuromuscular disease, hemophilia, immunodeficiencies, and cancer. Dunbar et al. review the pioneering work that led the gene therapy field to its current state, describe gene-editing technologies that are expected to play a major role in the field's future, and discuss practical challenges in getting these therapies to patients who need them. Science, this issue p. eaan4672 BACKGROUND Nearly five decades ago, visionary scientists hypothesized that genetic modification by exogenous DNA might be an effective treatment for inherited human diseases. This “gene therapy” strategy offered the theoretical advantage that a durable and possibly curative clinical benefit would be achieved by a single treatment. Although the journey from concept to clinical application has been long and tortuous, gene therapy is now bringing new treatment options to multiple fields of medicine. We review critical discoveries leading to the development of successful gene therapies, focusing on direct in vivo administration of viral vectors, adoptive transfer of genetically engineered T cells or hematopoietic stem cells, and emerging genome editing technologies. ADVANCES The development of gene delivery vectors such as replication-defective retro viruses and adeno-associated virus (AAV), coupled with encouraging results in preclinical disease models, led to the initiation of clinical trials in the early 1990s. Unfortunately, these early trials exposed serious therapy-related toxicities, including inflammatory responses to the vectors and malignancies caused by vector-mediated insertional activation of proto-oncogenes. These setbacks fueled more basic research in virology, immunology, cell biology, model development, and target disease, which ultimately led to successful clinical translation of gene therapies in the 2000s. Lentiviral vectors improved efficiency of gene transfer to nondividing cells. In early-phase clinical trials, these safer and more efficient vectors were used for transduction of autologous hematopoietic stem cells, leading to clinical benefit in patients with immunodeficiencies, hemoglobinopathies, and metabolic and storage disorders. T cells engineered to express CD19-specific chimeric antigen receptors were shown to have potent antitumor activity in patients with lymphoid malignancies. In vivo delivery of therapeutic AAV vectors to the retina, liver, and nervous system resulted in clinical improvement in patients with congenital blindness, hemophilia B, and spinal muscular atrophy, respectively. In the United States, Food and Drug Administration (FDA) approvals of the first gene therapy products occurred in 2017, including chimeric antigen receptor (CAR)–T cells to treat B cell malignancies and AAV vectors for in vivo treatment of congenital blindness. Promising clinical trial results in neuromuscular diseases and hemophilia will likely result in additional approvals in the near future. In recent years, genome editing technologies have been developed that are based on engineered or bacterial nucleases. In contrast to viral vectors, which can mediate only gene addition, genome editing approaches offer a precise scalpel for gene addition, gene ablation, and gene “correction.” Genome editing can be performed on cells ex vivo or the editing machinery can be delivered in vivo to effect in situ genome editing. Translation of these technologies to patient care is in its infancy in comparison to viral gene addition therapies, but multiple clinical genome editing trials are expected to open over the next decade. OUTLOOK Building on decades of scientific, clinical, and manufacturing advances, gene therapies have begun to improve the lives of patients with cancer and a variety of inherited genetic diseases. Partnerships with biotechnology and pharmaceutical companies with expertise in manufacturing and scale-up will be required for these therapies to have a broad impact on human disease. Many challenges remain, including understanding and preventing genotoxicity from integrating vectors or off-target genome editing, improving gene transfer or editing efficiency to levels necessary for treatment of many target diseases, preventing immune responses that limit in vivo administration of vectors or genome editing complexes, and overcoming manufacturing and regulatory hurdles. Importantly, a societal consensus must be reached on the ethics of germline genome editing in light of rapid scientific advances that have made this a real, rather than hypothetical, issue. Finally, payers and gene therapy clinicians and companies will need to work together to design and test new payment models to facilitate delivery of expensive but potentially curative therapies to patients in need. The ability of gene therapies to provide durable benefits to human health, exemplified by the scientific advances and clinical successes over the past several years, justifies continued optimism and increasing efforts toward making these therapies part of our standard treatment armamentarium for human disease. Three essential tools for human gene therapy. AAV and lentiviral vectors are the basis of several recently approved gene therapies. Gene editing technologies are in their translational and clinical infancy but are expected to play an increasing role in the field. After almost 30 years of promise tempered by setbacks, gene therapies are rapidly becoming a critical component of the therapeutic armamentarium for a variety of inherited and acquired human diseases. Gene therapies for inherited immune disorders, hemophilia, eye and neurodegenerative disorders, and lymphoid cancers recently progressed to approved drug status in the United States and Europe, or are anticipated to receive approval in the near future. In this Review, we discuss milestones in the development of gene therapies, focusing on direct in vivo administration of viral vectors and adoptive transfer of genetically engineered T cells or hematopoietic stem cells. We also discuss emerging genome editing technologies that should further advance the scope and efficacy of gene therapy approaches.

[1]  D. Perry,et al.  AAV5–Factor VIII Gene Transfer in Severe Hemophilia A , 2017, The New England journal of medicine.

[2]  J. Rasko,et al.  Hemophilia B Gene Therapy with a High‐Specific‐Activity Factor IX Variant , 2017, The New England journal of medicine.

[3]  Brian K. Kaspar,et al.  Single‐Dose Gene‐Replacement Therapy for Spinal Muscular Atrophy , 2017, The New England journal of medicine.

[4]  Rick L. Stevens,et al.  A communal catalogue reveals Earth’s multiscale microbial diversity , 2017, Nature.

[5]  David A. Williams,et al.  Hematopoietic Stem‐Cell Gene Therapy for Cerebral Adrenoleukodystrophy , 2017, The New England journal of medicine.

[6]  Kathleen A. Marshall,et al.  Efficacy and safety of voretigene neparvovec (AAV2-hRPE65v2) in patients with RPE65-mediated inherited retinal dystrophy: a randomised, controlled, open-label, phase 3 trial , 2017, The Lancet.

[7]  Jennifer A. Doudna,et al.  Enhanced proofreading governs CRISPR-Cas9 targeting accuracy , 2017, Nature.

[8]  K. Talbot,et al.  The clinical landscape for SMA in a new therapeutic era , 2017, Gene Therapy.

[9]  Michel Sadelain,et al.  Therapeutic T cell engineering , 2017, Nature.

[10]  Michel Sadelain,et al.  Chimeric Antigen Receptors: A Cell and Gene Therapy Perspective. , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[11]  G. Veres,et al.  Gene Therapy in a Patient with Sickle Cell Disease: Brief Report , 2017, The New England journal of medicine.

[12]  Mithat Gönen,et al.  Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection , 2017, Nature.

[13]  Adrian J. Thrasher,et al.  Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells , 2017, Science Translational Medicine.

[14]  David R. Liu,et al.  CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes , 2017, Cell.

[15]  Pachai Natarajan,et al.  CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease , 2017, Science Translational Medicine.

[16]  S. Gill Chimeric antigen receptor T cell therapy in AML: How close are we? , 2016, Best practice & research. Clinical haematology.

[17]  Syed Abbas Ali,et al.  T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. , 2016, Blood.

[18]  I. Verma,et al.  Lentiviral vectors, two decades later , 2016, Science.

[19]  S. Heimfeld,et al.  Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor–modified T cells , 2016, Science Translational Medicine.

[20]  Prashant Mali,et al.  A multifunctional AAV–CRISPR–Cas9 and its host response , 2016, Nature Methods.

[21]  R. Hardison,et al.  A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition , 2016, Nature Medicine.

[22]  Kathleen A. Marshall,et al.  Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial , 2016, The Lancet.

[23]  P. Rancoita,et al.  Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: an ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial , 2016, The Lancet.

[24]  A. Lanzavecchia,et al.  Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease , 2016, Science.

[25]  S. Orkin,et al.  Paying for future success in gene therapy , 2016, Science.

[26]  L. Notarangelo,et al.  Lentiviral hematopoietic stem cell gene therapy for X-linked severe combined immunodeficiency , 2016, Science Translational Medicine.

[27]  David R. Liu,et al.  Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage , 2016, Nature.

[28]  J. Joung,et al.  Defining and improving the genome-wide specificities of CRISPR–Cas9 nucleases , 2016, Nature Reviews Genetics.

[29]  X. Anguela,et al.  Adeno-associated viral vectors for the treatment of hemophilia. , 2016, Human molecular genetics.

[30]  M. Sadelain,et al.  Cell and Gene Therapy for the Beta-Thalassemias: Advances and Prospects. , 2016, Human gene therapy.

[31]  V. Buchholz,et al.  Role of memory T cell subsets for adoptive immunotherapy. , 2016, Seminars in immunology.

[32]  Dongsheng Duan,et al.  In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy , 2016, Science.

[33]  John M. Shelton,et al.  Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy , 2016, Science.

[34]  George M. Church,et al.  In vivo gene editing in dystrophic mouse muscle and muscle stem cells , 2016, Science.

[35]  S. Rosenberg,et al.  Prospects for gene-engineered T cell immunotherapy for solid cancers , 2016, Nature Medicine.

[36]  J. Joung,et al.  High-fidelity CRISPR-Cas9 variants with undetectable genome-wide off-targets , 2015, Nature.

[37]  David V Schaffer,et al.  Viral Vectors for Gene Therapy: Translational and Clinical Outlook. , 2015, Annual review of biomedical engineering.

[38]  David A. Scott,et al.  Rationally engineered Cas9 nucleases with improved specificity , 2015, Science.

[39]  Luigi Naldini,et al.  Gene therapy returns to centre stage , 2015, Nature.

[40]  Y. Doyon,et al.  In vivo genome editing of the albumin locus as a platform for protein replacement therapy. , 2015, Blood.

[41]  Matthew C. Canver,et al.  BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis , 2015, Nature.

[42]  Y. Kaneda,et al.  ASGCT and JSGT Joint Position Statement on Human Genomic Editing. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[43]  A. J. Roman,et al.  Improvement and decline in vision with gene therapy in childhood blindness. , 2015, New England Journal of Medicine.

[44]  S. E. Barker,et al.  Long-term effect of gene therapy on Leber's congenital amaurosis. , 2015, The New England journal of medicine.

[45]  A. Wright Long-term effects of retinal gene therapy in childhood blindness. , 2015, The New England journal of medicine.

[46]  Lei Zhang,et al.  Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. , 2015, Blood.

[47]  Frederic D Bushman,et al.  Outcomes following gene therapy in patients with severe Wiskott-Aldrich syndrome. , 2015, JAMA.

[48]  Ying Sun,et al.  CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes , 2015, Protein & Cell.

[49]  S. Riddell,et al.  Designing chimeric antigen receptors to effectively and safely target tumors. , 2015, Current opinion in immunology.

[50]  Sadik H. Kassim,et al.  Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[51]  S. Steinberg,et al.  T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial , 2015, The Lancet.

[52]  A. Miller Retroviral vectors: from cancer viruses to therapeutic tools. , 2014, Human gene therapy.

[53]  J. Doudna,et al.  The new frontier of genome engineering with CRISPR-Cas9 , 2014, Science.

[54]  M. Kay,et al.  Long-term safety and efficacy of factor IX gene therapy in hemophilia B. , 2014, The New England journal of medicine.

[55]  T. Ohmori,et al.  The prevalence of neutralizing antibodies against adeno‐associated virus capsids is reduced in young Japanese individuals , 2014, Journal of medical virology.

[56]  Pamela A Shaw,et al.  Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.

[57]  Christopher Baum,et al.  A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. , 2014, The New England journal of medicine.

[58]  S. Grupp,et al.  Current concepts in the diagnosis and management of cytokine release syndrome. , 2014, Blood.

[59]  Dana Carroll,et al.  Genome engineering with targetable nucleases. , 2014, Annual review of biochemistry.

[60]  M. van der Burg,et al.  Targeted Genome Editing in Human Repopulating Hematopoietic Stem Cells , 2014, Nature.

[61]  Michael Rothe,et al.  Gene Therapy for Wiskott-Aldrich Syndrome—Long-Term Efficacy and Genotoxicity , 2014, Science Translational Medicine.

[62]  Wei-Ting Hwang,et al.  Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. , 2014, The New England journal of medicine.

[63]  Jeffry D. Sander,et al.  CRISPR-Cas systems for editing, regulating and targeting genomes , 2014, Nature Biotechnology.

[64]  Z. Eshhar,et al.  The emergence of T-bodies/CAR T cells. , 2014, Cancer journal.

[65]  Qing He,et al.  Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia , 2014, Science Translational Medicine.

[66]  H. Ertl,et al.  Role of the vector genome and underlying factor IX mutation in immune responses to AAV gene therapy for hemophilia B , 2014, Journal of Translational Medicine.

[67]  Jennifer Couzin-Frankel,et al.  Breakthrough of the year 2013. Cancer immunotherapy. , 2013, Science.

[68]  C. von Kalle,et al.  Lentiviral Hematopoietic Stem Cell Gene Therapy Benefits Metachromatic Leukodystrophy , 2013, Science.

[69]  Luca Biasco,et al.  Lentiviral Hematopoietic Stem Cell Gene Therapy in Patients with Wiskott-Aldrich Syndrome , 2013, Science.

[70]  Luke A. Gilbert,et al.  CRISPR-Mediated Modular RNA-Guided Regulation of Transcription in Eukaryotes , 2013, Cell.

[71]  K. High,et al.  Immune responses to AAV vectors: overcoming barriers to successful gene therapy. , 2013, Blood.

[72]  P. Gregory,et al.  Genomic Editing of the HIV-1 Coreceptor CCR5 in Adult Hematopoietic Stem and Progenitor Cells Using Zinc Finger Nucleases , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[73]  Michel Sadelain,et al.  The basic principles of chimeric antigen receptor design. , 2013, Cancer discovery.

[74]  Qing He,et al.  CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia , 2013, Science Translational Medicine.

[75]  Luke A. Gilbert,et al.  Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression , 2013, Cell.

[76]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[77]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[78]  J. Keith Joung,et al.  TALENs: a widely applicable technology for targeted genome editing , 2012, Nature Reviews Molecular Cell Biology.

[79]  C. Klebanoff,et al.  Paths to stemness: building the ultimate antitumour T cell , 2012, Nature Reviews Cancer.

[80]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[81]  Karine Tremblay,et al.  Efficacy and long term safety of alipogene tiparvovec (AAV1-LPLS447X) gene therapy for lipoprotein lipase deficiency: an open label trial , 2012, Gene Therapy.

[82]  B. Byrne,et al.  Gene Therapy for Aromatic l-Amino Acid Decarboxylase Deficiency , 2012, Science Translational Medicine.

[83]  David V Schaffer,et al.  The AAV Vector Toolkit: Poised at the Clinical Crossroads. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[84]  Jeffry D. Sander,et al.  FLASH Assembly of TALENs Enables High-Throughput Genome Editing , 2012, Nature Biotechnology.

[85]  Chuanfeng Wu,et al.  Stem cell gene therapy: the risks of insertional mutagenesis and approaches to minimize genotoxicity , 2011, Frontiers of medicine.

[86]  Pratima Chowdary,et al.  Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. , 2011, The New England journal of medicine.

[87]  Michel Sadelain,et al.  Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. , 2011, Blood.

[88]  U. Modlich,et al.  Concise Review: Managing Genotoxicity in the Therapeutic Modification of Stem Cells , 2011, Stem cells.

[89]  A. Bagg,et al.  Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. , 2011, The New England journal of medicine.

[90]  J. F. Wright,et al.  Adeno-associated viral vector manufacturing: keeping pace with accelerating clinical development. , 2011, Human gene therapy.

[91]  A. Rezai,et al.  AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial , 2011, The Lancet Neurology.

[92]  M. Raffeld,et al.  Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[93]  P. Duchateau,et al.  Meganucleases and Other Tools for Targeted Genome Engineering: Perspectives and Challenges for Gene Therapy , 2011, Current gene therapy.

[94]  J. Jankovic,et al.  Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial , 2010, The Lancet Neurology.

[95]  W. Wilson,et al.  Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. , 2010, Blood.

[96]  Erin L. Doyle,et al.  Targeting DNA Double-Strand Breaks with TAL Effector Nucleases , 2010, Genetics.

[97]  Jérôme Larghero,et al.  Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia , 2010, Nature.

[98]  E. Rebar,et al.  Genome editing with engineered zinc finger nucleases , 2010, Nature Reviews Genetics.

[99]  H. Mizukami,et al.  A Phase I Study of Aromatic L-Amino Acid Decarboxylase Gene Therapy for Parkinson's Disease. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[100]  Sheng Huang,et al.  TAL nucleases (TALNs): hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain , 2010, Nucleic Acids Res..

[101]  Hans Martin,et al.  Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease , 2010, Nature Medicine.

[102]  Jens Boch,et al.  Breaking the Code of DNA Binding Specificity of TAL-Type III Effectors , 2009, Science.

[103]  Matthew J. Moscou,et al.  A Simple Cipher Governs DNA Recognition by TAL Effectors , 2009, Science.

[104]  Alessandro Aiuti,et al.  Gene therapy for immunodeficiency due to adenosine deaminase deficiency. , 2009, The New England journal of medicine.

[105]  W. Jagust,et al.  Safety and tolerability of putaminal AADC gene therapy for Parkinson disease , 2009, Neurology.

[106]  Kathleen A. Marshall,et al.  Age-dependent effects of RPE65 gene therapy for Leber's congenital amaurosis: a phase 1 dose-escalation trial , 2009, The Lancet.

[107]  Manfred Schmidt,et al.  Hematopoietic Stem Cell Gene Therapy with a Lentiviral Vector in X-Linked Adrenoleukodystrophy , 2009, Science.

[108]  Christof von Kalle,et al.  The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. , 2009, The Journal of clinical investigation.

[109]  R. Herzog,et al.  Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy. , 2009, Blood.

[110]  W. Hauswirth,et al.  Treatment of leber congenital amaurosis due to RPE65 mutations by ocular subretinal injection of adeno-associated virus gene vector: short-term results of a phase I trial. , 2008, Human gene therapy.

[111]  J. Orange,et al.  Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases , 2008, Nature Biotechnology.

[112]  N. Muzyczka,et al.  Next generation of adeno-associated virus 2 vectors: Point mutations in tyrosines lead to high-efficiency transduction at lower doses , 2008, Proceedings of the National Academy of Sciences.

[113]  Nick Tyler,et al.  Effect of gene therapy on visual function in Leber's congenital amaurosis. , 2008, The New England journal of medicine.

[114]  K. High,et al.  Immune responses to AAV in clinical trials. , 2007, Current gene therapy.

[115]  Marcela V Maus,et al.  CD8+ T-cell responses to adeno-associated virus capsid in humans , 2007, Nature Medicine.

[116]  A. Nienhuis,et al.  Genotoxicity of retroviral integration in hematopoietic cells. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[117]  J. Rasko,et al.  Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response , 2006, Nature Medicine.

[118]  D. Campana,et al.  Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia , 2004, Leukemia.

[119]  Cameron S. Osborne,et al.  LMO2-Associated Clonal T Cell Proliferation in Two Patients after Gene Therapy for SCID-X1 , 2003, Science.

[120]  Shawn M. Burgess,et al.  Transcription Start Regions in the Human Genome Are Favored Targets for MLV Integration , 2003, Science.

[121]  S. Larson,et al.  Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15 , 2003, Nature Medicine.

[122]  A. Mortellaro,et al.  Correction of ADA-SCID by Stem Cell Gene Therapy Combined with Nonmyeloablative Conditioning , 2002, Science.

[123]  A. Fischer,et al.  Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. , 2002, The New England journal of medicine.

[124]  R. Herzog,et al.  Sustained phenotypic correction of hemophilia B dogs with a factor IX null mutation by liver-directed gene therapy. , 2002, Blood.

[125]  M. Matsumura,et al.  Behavioral recovery in a primate model of Parkinson's disease by triple transduction of striatal cells with adeno-associated viral vectors expressing dopamine-synthesizing enzymes. , 2002, Human gene therapy.

[126]  Michel Sadelain,et al.  Therapeutic haemoglobin synthesis in β-thalassaemic mice expressing lentivirus-encoded human β-globin , 2000, Nature.

[127]  Alan McClelland,et al.  Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector , 2000, Nature Genetics.

[128]  S. Jenks Gene Therapy Death - "Everyone Has to Share in the Guilt". , 2000, Journal of the National Cancer Institute.

[129]  D. Kohn,et al.  Improved expression in hematopoietic and lymphoid cells in mice after transplantation of bone marrow transduced with a modified retroviral vector. , 1999, Blood.

[130]  Y. Korin,et al.  Progression to the G1b Phase of the Cell Cycle Is Required for Completion of Human Immunodeficiency Virus Type 1 Reverse Transcription in T Cells , 1998, Journal of Virology.

[131]  M. Sadelain,et al.  Recombinant retroviruses pseudotyped with the vesicular stomatitis virus G glycoprotein mediate both stable gene transfer and pseudotransduction in human peripheral blood lymphocytes. , 1997, Blood.

[132]  James M. Wilson,et al.  Stable gene transfer and expression of human blood coagulation factor IX after intramuscular injection of recombinant adeno-associated virus. , 1997, Proceedings of the National Academy of Sciences of the United States of America.

[133]  B. Byrne,et al.  Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[134]  R. Samulski,et al.  Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated virus vector , 1996, Journal of virology.

[135]  F. Gage,et al.  In Vivo Gene Delivery and Stable Transduction of Nondividing Cells by a Lentiviral Vector , 1996, Science.

[136]  D. Russell,et al.  Foamy virus vectors , 1996, Journal of virology.

[137]  R. Blaese,et al.  High-efficiency retroviral-mediated gene transfer into human and nonhuman primate peripheral blood lymphocytes. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[138]  R. Mulligan,et al.  Effects of retroviral vector design on expression of human adenosine deaminase in murine bone marrow transplant recipients engrafted with genetically modified cells. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[139]  P. Rouet,et al.  Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[140]  C. Bordignon,et al.  Peripheral Blood Lymphocytes as Target Cells of Retroviral Vector-Mediated Gene Transfer , 1994 .

[141]  J. Garcia,et al.  Construction and properties of retrovirus packaging cells based on gibbon ape leukemia virus , 1991, Journal of virology.

[142]  R. Mulligan,et al.  Safe and efficient generation of recombinant retroviruses with amphotropic and ecotropic host ranges. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[143]  P. Kantoff,et al.  Self-inactivating retroviral vectors designed for transfer of whole genes into mammalian cells. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[144]  David A. Williams,et al.  Introduction of new genetic material into pluripotent haematopoietic stem cells of the mouse , 1984, Nature.

[145]  H. Temin,et al.  Construction of a helper cell line for avian reticuloendotheliosis virus cloning vectors , 1983, Molecular and cellular biology.

[146]  D. Baltimore,et al.  Construction of a retrovirus packaging mutant and its use to produce helper-free defective retrovirus , 1983, Cell.

[147]  T. Friedmann,et al.  Gene Therapy for Human Genetic Disease? , 1972, Science.

[148]  Michel Sadelain,et al.  Targeting tumours with genetically enhanced T lymphocytes , 2003, Nature Reviews Cancer.

[149]  Michel Sadelain,et al.  Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRζ /CD28 receptor , 2002, Nature Biotechnology.

[150]  C. Dunbar,et al.  Gene transfer to hematopoietic stem cells: implications for gene therapy of human disease. , 1996, Annual review of medicine.