Cell and Gene Therapy for the Beta-Thalassemias: Advances and Prospects.

The beta-thalassemias are inherited anemias caused by mutations that severely reduce or abolish expression of the beta-globin gene. Like sickle cell disease, a related beta-globin gene disorder, they are ideal candidates for performing a genetic correction in patient hematopoietic stem cells (HSCs). The most advanced approach utilizes complex lentiviral vectors encoding the human β-globin gene, as first reported by May et al. in 2000. Considerable progress toward the clinical implementation of this approach has been made in the past five years, based on effective CD34+ cell mobilization and improved lentiviral vector manufacturing. Four trials have been initiated in the United States and Europe. Of 16 evaluable subjects, 6 have achieved transfusion independence. One of them developed a durable clonal expansion, which regressed after several years without transformation. Although globin lentiviral vectors have so far proven to be safe, this occurrence suggests that powerful insulators with robust enhancer-blocking activity will further enhance this approach. The combined discovery of Bcl11a-mediated γ-globin gene silencing and advances in gene editing are the foundations for another gene therapy approach, which aims to reactivate fetal hemoglobin (HbF) production. Its clinical translation will hinge on the safety and efficiency of gene targeting in true HSCs and the induction of sufficient levels of HbF to achieve transfusion independence. Altogether, the progress achieved over the past 15 years bodes well for finding a genetic cure for severe globin disorders in the next decade.

[1]  Jean-Paul Concordet,et al.  Improved Genome Editing Efficiency and Flexibility Using Modified Oligonucleotides with TALEN and CRISPR-Cas9 Nucleases. , 2016, Cell reports.

[2]  Dana Carroll,et al.  Origins of Programmable Nucleases for Genome Engineering. , 2016, Journal of molecular biology.

[3]  Isaac B. Hilton,et al.  Editing the epigenome: technologies for programmable transcription and epigenetic modulation , 2016, Nature Methods.

[4]  S. Borwornpinyo,et al.  Gene Therapy of the β-Hemoglobinopathies by Lentiviral Transfer of the βA(T87Q)-Globin Gene , 2016, Human gene therapy.

[5]  Morgan L. Maeder,et al.  Genome-editing Technologies for Gene and Cell Therapy , 2016, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  S. Yamanaka,et al.  From Genomics to Gene Therapy: Induced Pluripotent Stem Cells Meet Genome Editing. , 2015, Annual review of genetics.

[7]  Peter C. Fineran,et al.  The Cpf1 CRISPR-Cas protein expands genome-editing tools , 2015, Genome Biology.

[8]  P. Gregory,et al.  Homology-driven genome editing in hematopoietic stem and progenitor cells using zinc finger nuclease mRNA and AAV6 donors , 2015, Nature Biotechnology.

[9]  P. Gee,et al.  Minimizing off-Target Mutagenesis Risks Caused by Programmable Nucleases , 2015, International journal of molecular sciences.

[10]  A. Scharenberg,et al.  Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template , 2015, Science Translational Medicine.

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

[12]  Yuri R. Bendaña,et al.  Functional footprinting of regulatory DNA , 2015, Nature Methods.

[13]  S. Orkin,et al.  Hemoglobin switching's surprise: the versatile transcription factor BCL11A is a master repressor of fetal hemoglobin. , 2015, Current opinion in genetics & development.

[14]  De-Pei Liu,et al.  Both TALENs and CRISPR/Cas9 directly target the HBB IVS2–654 (C > T) mutation in β-thalassemia-derived iPSCs , 2015, Scientific Reports.

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

[16]  Christopher M. Vockley,et al.  Epigenome editing by a CRISPR/Cas9-based acetyltransferase activates genes from promoters and enhancers , 2015, Nature Biotechnology.

[17]  Manfred Schmidt,et al.  Mapping the precision of genome editing , 2015, Nature Biotechnology.

[18]  Matthew T. Maurano,et al.  Genomic discovery of potent chromatin insulators for human gene therapy , 2015, Nature Biotechnology.

[19]  Y. Kan,et al.  Seamless gene correction of β-thalassemia mutations in patient-specific iPSCs using CRISPR/Cas9 and piggyBac , 2014, Genome research.

[20]  Philip D. Gregory,et al.  Reactivation of Developmentally Silenced Globin Genes by Forced Chromatin Looping , 2014, Cell.

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

[22]  H. Kim,et al.  A guide to genome engineering with programmable nucleases , 2014, Nature Reviews Genetics.

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

[24]  M. Sadelain,et al.  Safe mobilization of CD34+ cells in adults with β-thalassemia and validation of effective globin gene transfer for clinical investigation. , 2014, Blood.

[25]  Rodney Rothstein,et al.  Repair of strand breaks by homologous recombination. , 2013, Cold Spring Harbor perspectives in biology.

[26]  Shondra M. Pruett-Miller,et al.  Nuclease-mediated gene editing by homologous recombination of the human globin locus , 2013, Nucleic acids research.

[27]  Hui Zhang,et al.  Transcription Activator-like Effector Nuclease (TALEN)-mediated Gene Correction in Integration-free β-Thalassemia Induced Pluripotent Stem Cells* , 2013, The Journal of Biological Chemistry.

[28]  Matthew C. Canver,et al.  An Erythroid Enhancer of BCL11A Subject to Genetic Variation Determines Fetal Hemoglobin Level , 2013, Science.

[29]  Christopher M. Vockley,et al.  RNA-guided gene activation by CRISPR-Cas9-based transcription factors , 2013, Nature Methods.

[30]  Jennifer E. Phillips-Cremins,et al.  Chromatin insulators: linking genome organization to cellular function. , 2013, Molecular cell.

[31]  P. Glazer,et al.  Systemic delivery of triplex-forming PNA and donor DNA by nanoparticles mediates site-specific genome editing of human hematopoietic cells in vivo , 2012, Gene Therapy.

[32]  Morgan L. Maeder,et al.  In Situ Genetic Correction of the Sickle Cell Anemia Mutation in Human Induced Pluripotent Stem Cells Using Engineered Zinc Finger Nucleases , 2011, Stem cells.

[33]  P. Mali,et al.  Site-specific gene correction of a point mutation in human iPS cells derived from an adult patient with sickle cell disease. , 2011, Blood.

[34]  A. Kattamis,et al.  Survival in a large cohort of Greek patients with transfusion‐dependent beta thalassaemia and mortality ratios compared to the general population , 2011, European journal of haematology.

[35]  Shoshannah L. Roth,et al.  A method to sequence and quantify DNA integration for monitoring outcome in gene therapy , 2011, Nucleic acids research.

[36]  D. W. Emery The use of chromatin insulators to improve the expression and safety of integrating gene transfer vectors. , 2011, Human gene therapy.

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

[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]  F. Warburton,et al.  Improved survival in thalassemia major patients on switching from desferrioxamine to combined chelation therapy with desferrioxamine and deferiprone , 2009, Haematologica.

[40]  Stuart H. Orkin,et al.  Developmental and species-divergent globin switching are driven by BCL11A , 2009, Nature.

[41]  M. Sadelain,et al.  Stem cell engineering for the treatment of severe hemoglobinopathies. , 2008, Current molecular medicine.

[42]  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.

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

[44]  J. Hirschhorn,et al.  DNA polymorphisms at the BCL11A, HBS1L-MYB, and β-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease , 2008, Proceedings of the National Academy of Sciences.

[45]  A. Miccio,et al.  In vivo selection of genetically modified erythroblastic progenitors leads to long-term correction of β-thalassemia , 2008, Proceedings of the National Academy of Sciences.

[46]  Gonçalo R. Abecasis,et al.  Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of β-thalassemia , 2008, Proceedings of the National Academy of Sciences.

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

[48]  Simon Heath,et al.  A QTL influencing F cell production maps to a gene encoding a zinc-finger protein on chromosome 2p15 , 2007, Nature Genetics.

[49]  M. Sadelain,et al.  The genetic engineering of hematopoietic stem cells: the rise of lentiviral vectors, the conundrum of the ltr, and the promise of lineage-restricted vectors. , 2007, Molecular therapy : the journal of the American Society of Gene Therapy.

[50]  S. Rivella,et al.  Therapeutic options for patients with severe beta-thalassemia: the need for globin gene therapy. , 2006, Human gene therapy.

[51]  M. Sadelain Recent advances in globin gene transfer for the treatment of beta-thalassemia and sickle cell anemia , 2006, Current opinion in hematology.

[52]  Yang Du,et al.  Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1 , 2006, Nature Medicine.

[53]  A. Mancuso,et al.  A Prospective Study of Hepatocellular Carcinoma Incidence in Thalassemia , 2006, Hemoglobin.

[54]  P. Leboulch,et al.  A Phase I/II Clinical Trial of β‐Globin Gene Therapy for β‐Thalassemia , 2005 .

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

[56]  A. Nienhuis,et al.  Gene therapy for the hemoglobin disorders. , 2003, Current hematology reports.

[57]  M. Sadelain,et al.  Occurrence of leukaemia following gene therapy of X-linked SCID , 2003, Nature Reviews Cancer.

[58]  S. Rivella,et al.  A novel murine model of Cooley anemia and its rescue by lentiviral-mediated human beta-globin gene transfer. , 2003, Blood.

[59]  Xiangdong Fang,et al.  Locus control regions. , 2002, Blood.

[60]  G. Stamatoyannopoulos,et al.  Development of virus vectors for gene therapy of beta chain hemoglobinopathies: flanking with a chromatin insulator reduces gamma-globin gene silencing in vivo. , 2002, Blood.

[61]  S. Rivella,et al.  Successful treatment of murine β-thalassemia intermedia by transfer of the human β-globin gene , 2002 .

[62]  G. Stamatoyannopoulos,et al.  A chromatin insulator protects retrovirus vectors from chromosomal position effects. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

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

[64]  S. Rivella,et al.  The cHS4 Insulator Increases the Probability of Retroviral Expression at Random Chromosomal Integration Sites , 2000, Journal of Virology.

[65]  K. Li,et al.  Granulocyte colony–stimulating factor–mobilized peripheral blood stem cells in β-thalassemia patients: kinetics of mobilization and composition of apheresis product , 1999 .

[66]  B. Forget Molecular Basis of Hereditary Persistence of Fetal Hemoglobin , 1998, Annals of the New York Academy of Sciences.

[67]  P. Rouet,et al.  Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. , 1994, Molecular and cellular biology.

[68]  D. Labie,et al.  Structural bases of the inhibitory effects of hemoglobin F and hemoglobin A2 on the polymerization of hemoglobin S. , 1979, Proceedings of the National Academy of Sciences of the United States of America.

[69]  A. D’Andrea,et al.  Repair Pathway Choices and Consequences at the Double-Strand Break. , 2016, Trends in cell biology.

[70]  G. Stamatoyannopoulos,et al.  Hematopoietic stem cell mobilization for gene therapy of adult patients with severe β-thalassemia: results of clinical trials using G-CSF or plerixafor in splenectomized and nonsplenectomized subjects. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[71]  R. Ghirlando,et al.  Chromatin boundaries, insulators, and long-range interactions in the nucleus. , 2010, Cold Spring Harbor symposia on quantitative biology.

[72]  Yusuke Nakamura,et al.  A genome-wide association identified the common genetic variants influence disease severity in β0-thalassemia/hemoglobin E , 2009, Human Genetics.

[73]  A. Cnaan,et al.  Survival and complications in patients with thalassemia major treated with transfusion and deferoxamine. , 2004, Haematologica.

[74]  J. Hirschhorn,et al.  Supporting Online Material Materials and Methods Figs. S1 to S10 Tables S1 to S7 References Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-specific Repressor Bcl11a , 2022 .