Lentiviral Hematopoietic Stem Cell Gene Therapy in Patients with Wiskott-Aldrich Syndrome

Introduction Wiskott-Aldrich syndrome (WAS) is a primary immunodeficiency characterized by eczema, thrombocytopenia, infections, and a high risk of developing autoimmunity and cancer. In a recent clinical trial, a γ-retroviral vector was used to introduce a functional WAS gene into autologous hematopoietic stem/progenitor cells (HSCs), followed by reinfusion of the gene-corrected HSCs into the patients. This strategy provided clinical benefit but resulted in expansion and malignant transformation of hematopoietic clones carrying vector insertions near oncogenes, thus increasing leukemia risk. We have developed a clinical protocol for WAS based on lentiviral vector (LV) gene transfer into HSCs. Analysis of common insertion sites (CIS) in gene therapy trials using lentiviral versus γ-retroviral vectors. Word clouds show the intensity of insertion sites clustering in each of the CIS genes (the larger the gene name, the larger the number of insertion sites within or in the proximity of that gene). The names of the CIS genes detected in both gene therapy trials are reported at the intersection between the circles. Analysis of common insertion sites (CIS) in gene therapy trials using lentiviral versus γ-retroviral vectors. Word clouds show the intensity of insertion sites clustering in each of the CIS genes (the larger the gene name, the larger the number of insertion sites within or in the proximity of that gene). The names of the CIS genes detected in both gene therapy trials are reported at the intersection between the circles. Methods Three patients with WAS were treated in a phase I/II clinical trial with gene-corrected HSCs after pretreatment with a reduced-intensity myeloablative regimen. Autologous CD34+ cells were transduced with an optimized LV carrying the WAS gene under the control of its endogenous promoter. Patients were monitored for up to 2.5 years after gene therapy by molecular, immunological, and clinical tests. We also investigated the genomic distribution of LV integration sites in the patients’ bone marrow and peripheral blood cell lineages. Results Administration of autologous HSCs transduced with LV at high efficiency (>90%) resulted in robust (25 to 50%), stable, and long-term engraftment of gene-corrected HSCs in the patients’ bone marrow. WAS protein expression was detected in myeloid cells at similar rates and in nearly all circulating platelets and lymphoid cells. In vitro T cell proliferative responses, natural killer cell cytotoxic activity, immune synapsis formation, and suppressive function of T regulatory cells were normalized. In all three patients, we observed improved platelet counts, protection from bleeding and severe infections, and resolution of eczema. Vector integration analyses on >35,000 unique insertion sites showed distinct waves of HSC clonal output, resulting in highly polyclonal multilineage hematopoietic reconstitution. In contrast to ©-retroviral gene therapy, our LV-based therapy did not induce in vivo selection of clones carrying integrations near oncogenes. Consistent with this, we did not see evidence of clonal expansions in the patients for up to 20 to 32 months after gene therapy. Discussion Our gene transfer protocol provided efficient stem cell transduction in vitro, resulting in robust and stable in vivo gene marking. WAS expression was restored to near-physiological levels in the patients, resulting in immunological and hematological improvement and clinical benefit. Clonal tracking of stem cell dynamics by vector insertions showed details of hematopoietic reconstitution after gene therapy. Comparison with clinical data from ©-retroviral gene therapy in the same disease setting strongly suggests that LV gene therapy offers safety advantages, but a longer follow-up time is needed for validation. Collectively, our findings support the use of LV gene therapy to treat patients with WAS and other hematological disorders. Next-Generation Gene Therapy Few disciplines in contemporary clinical research have experienced the high expectations directed at the gene therapy field. However, gene therapy has been challenging to translate to the clinic, often because the therapeutic gene is expressed at insufficient levels in the patient or because the gene delivery vector integrates near protooncogenes, which can cause leukemia (see the Perspective by Verma). Biffi et al. (1233158, published online 11 July) and Aiuti et al. (1233151; published online 11 July) report progress on both fronts in gene therapy trials of three patients with metachromatic leukodystrophy (MLD), a neurodegenerative disorder, and three patients with Wiskott-Aldrich syndrome (WAS), an immunodeficiency disorder. Optimized lentiviral vectors were used to introduce functional MLD or WAS genes into the patients' hematopoietic stem cells (HSCs) ex vivo, and the transduced cells were then infused back into the patients, who were then monitored for up to 2 years. In both trials, the patients showed stable engraftment of the transduced HSC and high expression levels of functional MLD or WAS genes. Encouragingly, there was no evidence of lentiviral vector integration near proto-oncogenes, and the gene therapy treatment halted disease progression in most patients. A longer follow-up period will be needed to further validate efficacy and safety. Lentivirus-mediated gene therapy produces encouraging results in three children with a rare immunodeficiency disorder. [Also see Perspective by Verma] Wiskott-Aldrich syndrome (WAS) is an inherited immunodeficiency caused by mutations in the gene encoding WASP, a protein regulating the cytoskeleton. Hematopoietic stem/progenitor cell (HSPC) transplants can be curative, but, when matched donors are unavailable, infusion of autologous HSPCs modified ex vivo by gene therapy is an alternative approach. We used a lentiviral vector encoding functional WASP to genetically correct HSPCs from three WAS patients and reinfused the cells after a reduced-intensity conditioning regimen. All three patients showed stable engraftment of WASP-expressing cells and improvements in platelet counts, immune functions, and clinical scores. Vector integration analyses revealed highly polyclonal and multilineage haematopoiesis resulting from the gene-corrected HSPCs. Lentiviral gene therapy did not induce selection of integrations near oncogenes, and no aberrant clonal expansion was observed after 20 to 32 months. Although extended clinical observation is required to establish long-term safety, lentiviral gene therapy represents a promising treatment for WAS.

[1]  T. Laín de Lera,et al.  Lentiviral vector-mediated gene transfer in T cells from Wiskott-Aldrich syndrome patients leads to functional correction. , 2004, Molecular therapy : the journal of the American Society of Gene Therapy.

[2]  H. Ochs,et al.  Wiskott-Aldrich syndrome: diagnosis, clinical and laboratory manifestations, and treatment. , 2009, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[3]  L. Notarangelo,et al.  Wiskott-Aldrich syndrome , 2008, Current opinion in hematology.

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

[5]  Kathryn L. Parsley,et al.  Long-Term Persistence of a Polyclonal T Cell Repertoire After Gene Therapy for X-Linked Severe Combined Immunodeficiency , 2011, Science Translational Medicine.

[6]  R. Day,et al.  Evaluation of tetrazolium-based semiautomatic colorimetric assay for measurement of human antitumor cytotoxicity. , 1990, Cancer research.

[7]  Clelia Di Serio,et al.  Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration , 2006, Nature Biotechnology.

[8]  Yasuhiko Ito,et al.  Mixed chimera status of 12 patients with Wiskott-Aldrich syndrome (WAS) after hematopoietic stem cell transplantation: evaluation by flow cytometric analysis of intracellular WAS protein expression. , 2002, Blood.

[9]  F. Candotti,et al.  Second-site mutation in the Wiskott-Aldrich syndrome (WAS) protein gene causes somatic mosaicism in two WAS siblings. , 2003, The Journal of clinical investigation.

[10]  L. Biasco,et al.  Retroviral Integrations in Gene Therapy Trials , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

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

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

[13]  F. Bushman,et al.  HIV integration site selection: analysis by massively parallel pyrosequencing reveals association with epigenetic modifications. , 2007, Genome research.

[14]  O. Danos,et al.  Lentiviral vectors targeting WASp expression to hematopoietic cells, efficiently transduce and correct cells from WAS patients , 2007, Gene Therapy.

[15]  L. Naldini Ex vivo gene transfer and correction for cell-based therapies , 2011, Nature Reviews Genetics.

[16]  F. Bushman,et al.  Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. , 2008, The Journal of clinical investigation.

[17]  Aaron R Cooper,et al.  Gene therapy for adenosine deaminase-deficient severe combined immune deficiency: clinical comparison of retroviral vectors and treatment plans. , 2012, Blood.

[18]  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.

[19]  Dustin E. Schones,et al.  Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. , 2009, Cell stem cell.

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

[21]  S. Kanner,et al.  Wiskott-Aldrich syndrome/X-linked thrombocytopenia: WASP gene mutations, protein expression, and phenotype. , 1997, Blood.

[22]  Anne Chao,et al.  An overview of closed capture-recapture models , 2001 .

[23]  C. von Kalle,et al.  Insertion sites in engrafted cells cluster within a limited repertoire of genomic areas after gammaretroviral vector gene therapy. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[24]  Gianluigi Zanetti,et al.  Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. , 2011, Blood.

[25]  L. Notarangelo,et al.  Defective Th1 Cytokine Gene Transcription in CD4+ and CD8+ T Cells from Wiskott-Aldrich Syndrome Patients1 , 2006, The Journal of Immunology.

[26]  Christof von Kalle,et al.  Stem-cell gene therapy for the Wiskott-Aldrich syndrome. , 2010, The New England journal of medicine.

[27]  F. Alt,et al.  Wiskott-Aldrich syndrome protein (WASP) and N-WASP are critical for peripheral B-cell development and function. , 2012, Blood.

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

[29]  Christof von Kalle,et al.  Analyzing the Number of Common Integration Sites of Viral Vectors – New Methods and Computer Programs , 2011, PloS one.

[30]  S. Davies,et al.  Outcomes following hematopoietic cell transplantation for Wiskott–Aldrich syndrome , 2012, Bone Marrow Transplantation.

[31]  Alessandro Aiuti,et al.  Hot spots of retroviral integration in human CD34+ hematopoietic cells. , 2007, Blood.

[32]  L. Naldini,et al.  Efficacy of gene therapy for Wiskott-Aldrich syndrome using a WAS promoter/cDNA-containing lentiviral vector and nonlethal irradiation. , 2006, Human gene therapy.

[33]  Stephan Wolf,et al.  Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing , 2010, Nature Protocols.

[34]  A. Fischer,et al.  Gene therapy for primary immunodeficiencies: Part 1. , 2012, Current opinion in immunology.

[35]  Hanno Glimm,et al.  High-resolution insertion-site analysis by linear amplification–mediated PCR (LAM-PCR) , 2007, Nature Methods.

[36]  A. Villa,et al.  Recent advances in understanding the pathophysiology of Wiskott-Aldrich syndrome. , 2009, Blood.

[37]  A. Fischer,et al.  Long-term outcome and lineage-specific chimerism in 194 patients with Wiskott-Aldrich syndrome treated by hematopoietic cell transplantation in the period 1980-2009: an international collaborative study. , 2011, Blood.

[38]  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.

[39]  M. Roncarolo,et al.  Alterations in the adenosine metabolism and CD39/CD73 adenosinergic machinery cause loss of Treg cell function and autoimmunity in ADA-deficient SCID. , 2012, Blood.

[40]  S. Snapper,et al.  The Wiskott-Aldrich syndrome protein is required for the function of CD4+CD25+Foxp3+ regulatory T cells , 2007, The Journal of experimental medicine.

[41]  Alessandro Guffanti,et al.  High-definition mapping of retroviral integration sites identifies active regulatory elements in human multipotent hematopoietic progenitors. , 2010, Blood.

[42]  M. C. Castiello,et al.  Autoimmunity in Wiskott–Aldrich Syndrome: An Unsolved Enigma , 2012, Front. Immun..

[43]  Christine Kinnon,et al.  Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. , 2008, The Journal of clinical investigation.

[44]  Gerald de Haan,et al.  Counting stem cells: methodological constraints , 2012, Nature Methods.

[45]  L. Naldini,et al.  Lentiviral-mediated gene therapy leads to improvement of B-cell functionality in a murine model of Wiskott-Aldrich syndrome. , 2011, The Journal of allergy and clinical immunology.

[46]  J. Orange,et al.  IL-2 induces a WAVE2-dependent pathway for actin reorganization that enables WASp-independent human NK cell function. , 2011, The Journal of clinical investigation.

[47]  K. Siminovitch,et al.  Evidence for long-term efficacy and safety of gene therapy for Wiskott-Aldrich syndrome in preclinical models. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[48]  H. Ochs,et al.  Ubiquitous high-level gene expression in hematopoietic lineages provides effective lentiviral gene therapy of murine Wiskott-Aldrich syndrome. , 2012, Blood.

[49]  Frederic D. Bushman,et al.  Efficacy of gene therapy for X-linked severe combined immunodeficiency. , 2010, The New England journal of medicine.

[50]  A. Schambach,et al.  Insertional transformation of hematopoietic cells by self-inactivating lentiviral and gammaretroviral vectors. , 2009, Molecular therapy : the journal of the American Society of Gene Therapy.

[51]  Frederic D. Bushman,et al.  Dynamics of gene-modified progenitor cells analyzed by tracking retroviral integration sites in a human SCID-X1 gene therapy trial. , 2010, Blood.

[52]  L. Notarangelo,et al.  How I treat ADA deficiency. , 2009, Blood.

[53]  Hung Fan,et al.  Challenges in vector and trial design using retroviral vectors for long-term gene correction in hematopoietic stem cell gene therapy. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[54]  M. Cavazzana‐Calvo,et al.  Gene therapy for primary immunodeficiencies: Part 2. , 2012, Current opinion in immunology.

[55]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[56]  C. von Kalle,et al.  Preclinical Safety and Efficacy of Human CD34+ Cells Transduced With Lentiviral Vector for the Treatment of Wiskott-Aldrich Syndrome , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[57]  Michael J. Byrne,et al.  Wiskott–Aldrich syndrome protein is required for NK cell cytotoxicity and colocalizes with actin to NK cell-activating immunologic synapses , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[58]  Luca Biasco,et al.  Integration profile of retroviral vector in gene therapy treated patients is cell-specific according to gene expression and chromatin conformation of target cell , 2011, EMBO molecular medicine.

[59]  L. Seymour,et al.  Gene therapy matures in the clinic , 2012, Nature Biotechnology.

[60]  A. Fischer,et al.  Long-term outcome following hematopoietic stem-cell transplantation in Wiskott-Aldrich syndrome: collaborative study of the European Society for Immunodeficiencies and European Group for Blood and Marrow Transplantation. , 2008, Blood.

[61]  L. Notarangelo,et al.  WASP regulates suppressor activity of human and murine CD4+CD25+FOXP3+ natural regulatory T cells , 2007, The Journal of experimental medicine.