Genetic therapies for the first molecular disease.

Sickle cell disease (SCD) is a monogenic disorder characterized by recurrent episodes of severe bone pain, multi-organ failure, and early mortality. Although medical progress over the past several decades has improved clinical outcomes and offered cures for many affected individuals living in high-income countries, most SCD patients still experience substantial morbidity and premature death. Emerging technologies to manipulate somatic cell genomes and insights into the mechanisms of developmental globin gene regulation are generating potentially transformative approaches to cure SCD by autologous hematopoietic stem cell (HSC) transplantation. Key components of current approaches include ethical informed consent, isolation of patient HSCs, in vitro genetic modification of HSCs to correct the SCD mutation or circumvent its damaging effects, and reinfusion of the modified HSCs following myelotoxic bone marrow conditioning. Successful integration of these components into effective therapies requires interdisciplinary collaborations between laboratory researchers, clinical caregivers, and patients. Here we summarize current knowledge and research challenges for each key component, emphasizing that the best approaches have yet to be developed.

[1]  J. Mehta CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia. , 2021, The New England journal of medicine.

[2]  J. Tisdale,et al.  A pause in gene therapy: Reflecting on the unique challenges of sickle cell disease. , 2021, Molecular therapy : the journal of the American Society of Gene Therapy.

[3]  A. Wilkinson,et al.  Cas9-AAV6 gene correction of beta-globin in autologous HSCs improves sickle cell disease erythropoiesis in mice , 2020, Nature Communications.

[4]  David A. Williams,et al.  Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. , 2020, The New England journal of medicine.

[5]  J. Makani,et al.  The role of haematopoietic stem cell transplantation for sickle cell disease in the era of targeted disease-modifying therapies and gene editing. , 2020, The Lancet. Haematology.

[6]  David R. Liu,et al.  Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression , 2020, bioRxiv.

[7]  Nicole M. Gaudelli,et al.  Adenine Base Editing of the Sickle Allele in CD34+ Hematopoietic Stem and Progenitor Cells Eliminates Hemoglobin S , 2020 .

[8]  David R. Liu,et al.  Adenosine Base Editing of γ-Globin Promoters Induces Fetal Hemoglobin and Inhibit Erythroid Sickling , 2020, Blood.

[9]  David R. Liu,et al.  Base Editing Eliminates the Sickle Cell Mutation and Pathology in Hematopoietic Stem Cells Derived Erythroid Cells , 2020 .

[10]  S. Grupp,et al.  Safety and Efficacy of CTX001 in Patients with Transfusion-Dependent β-Thalassemia and Sickle Cell Disease: Early Results from the Climb THAL-111 and Climb SCD-121 Studies of Autologous CRISPR-CAS9-Modified CD34+ Hematopoietic Stem and Progenitor Cells , 2020 .

[11]  J. Rasko,et al.  Long-Term Efficacy and Safety of Betibeglogene Autotemcel Gene Therapy for the Treatment of Transfusion-Dependent β-Thalassemia: Results in Patients with up to 6 Years of Follow-up , 2020 .

[12]  Umut A. Gurkan,et al.  Standardized microfluidic assessment of red blood cell–mediated microcapillary occlusion: Association with clinical phenotype and hydroxyurea responsiveness in sickle cell disease , 2020, Microcirculation.

[13]  Yong-Sam Kim,et al.  Unbiased investigation of specificities of prime editing systems in human cells , 2020, Nucleic acids research.

[14]  A. Adekile The Genetic and Clinical Significance of Fetal Hemoglobin Expression in Sickle Cell Disease , 2020, Medical Principles and Practice.

[15]  Han Yang,et al.  Methods Favoring Homology-Directed Repair Choice in Response to CRISPR/Cas9 Induced-Double Strand Breaks , 2020, International journal of molecular sciences.

[16]  Markus S. Schröder,et al.  Controlled Cycling and Quiescence Enables Efficient HDR in Engraftment-Enriched Adult Hematopoietic Stem and Progenitor Cells , 2020, Cell reports.

[17]  M. Steinberg Fetal Hemoglobin in Sickle Cell Anemia. , 2020, Blood.

[18]  M. Firth,et al.  CRISPR GUARD protects off-target sites from Cas9 nuclease activity using short guide RNAs , 2020, Nature Communications.

[19]  A. Abate,et al.  Single cell mutation analysis of clonal evolution in myeloid malignancies , 2020, Nature.

[20]  Andrew T. Francis,et al.  Direct quantification of single red blood cell hemoglobin concentration with multiphoton microscopy. , 2020, Analytical chemistry.

[21]  L. Lai,et al.  AcrIIA5 Suppresses Base Editors and Reduces Their Off-Target Effects , 2020, Cells.

[22]  S. Bae,et al.  Current Status and Challenges of DNA Base Editing Tools. , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[23]  Yanpeng Wang,et al.  Rationally Designed APOBEC3B Cytosine Base Editors with Improved Specificity. , 2020, Molecular cell.

[24]  A. Kolomeisky,et al.  Single C-to-T substitution using engineered APOBEC3G-nCas9 base editors with minimum genome- and transcriptome-wide off-target effects , 2020, Science Advances.

[25]  Saman Majeed,et al.  Latest Developed Strategies to Minimize the Off-Target Effects in CRISPR-Cas-Mediated Genome Editing , 2020, Cells.

[26]  David R. Liu,et al.  Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors , 2020, Nature Biotechnology.

[27]  S. Tsang,et al.  Allele-Specific Chromosome Removal after Cas9 Cleavage in Human Embryos , 2020, Cell.

[28]  B. Le Pioufle,et al.  Characterization of red blood cell microcirculatory parameters using a bioimpedance microfluidic device , 2020, Scientific Reports.

[29]  M. Mokry,et al.  Prime editing for functional repair in patient-derived disease models , 2020, Nature Communications.

[30]  Oana M. Enache,et al.  Cas9 activates the p53 pathway and selects for p53-inactivating mutations , 2020, Nature Genetics.

[31]  Hui Yang,et al.  A rationally engineered cytosine base editor retains high on-target activity while reducing both DNA and RNA off-target effects , 2020, Nature Methods.

[32]  J. Tisdale,et al.  Myelodysplastic syndrome unrelated to lentiviral vector in a patient treated with gene therapy for sickle cell disease. , 2020, Blood advances.

[33]  Nicole M. Gaudelli,et al.  Cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity , 2020, Nature Communications.

[34]  B. Ryu,et al.  Optimizing lentiviral vector transduction of hematopoietic stem cells for gene therapy , 2020, Gene Therapy.

[35]  J. Tisdale,et al.  βT87Q-Globin Gene Therapy Reduces Sickle Hemoglobin Production, Allowing for Ex Vivo Anti-sickling Activity in Human Erythroid Cells , 2020, Molecular therapy. Methods & clinical development.

[36]  M. Pellegrini,et al.  Creating New β-Globin-Expressing Lentiviral Vectors by High-Resolution Mapping of Locus Control Region Enhancer Sequences , 2020, Molecular therapy. Methods & clinical development.

[37]  C. Grady,et al.  Motivations and decision-making of adult sickle cell patients in high-risk clinical research. , 2020, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[38]  David A. Williams,et al.  Preclinical Evaluation of a Novel Lentiviral Vector Driving Lineage-Specific BCL11A Knockdown for Sickle Cell Gene Therapy , 2020, Molecular therapy. Methods & clinical development.

[39]  C. Hourigan,et al.  Baseline TP53 mutations in Adults with SCD developing Myeloid Malignancy following Hematopoietic Cell Transplantation. , 2020, Blood.

[40]  Thomas E. Hughes,et al.  Safe and efficient peripheral blood stem cell collection in patients with sickle cell disease using plerixafor. , 2020, Haematologica.

[41]  F. Bushman,et al.  Clonal tracking in gene therapy patients reveals a diversity of human hematopoietic differentiation programs. , 2020, Blood.

[42]  J. Concordet,et al.  Editing a γ-globin repressor binding site restores fetal hemoglobin synthesis and corrects the sickle cell disease phenotype , 2020, Science Advances.

[43]  Jennifer A. Doudna,et al.  THE PROMISE AND CHALLENGE OF THERAPEUTIC GENOME EDITING , 2020, Nature.

[44]  Junjie Tan,et al.  Expanding the genome-targeting scope and the site selectivity of high-precision base editors , 2020, Nature Communications.

[45]  Charles P. Lin,et al.  Pathologic angiogenesis in the bone marrow of humanized sickle cell mice is reversible by blood transfusion. , 2020, Blood.

[46]  Fatih Kocabaş,et al.  Development of Gene Editing Strategies for Human β-Globin (HBB) Gene Mutations , 2020, bioRxiv.

[47]  David R. Liu,et al.  Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors , 2020, Nature Biotechnology.

[48]  Tony P. Huang,et al.  Continuous evolution of SpCas9 variants compatible with non-G PAMs , 2020, Nature Biotechnology.

[49]  A. Fischer,et al.  Gene therapy for severe combined immunodeficiencies and beyond , 2019, The Journal of experimental medicine.

[50]  Jason M. Gehrke,et al.  Therapeutic base editing of human hematopoietic stem cells , 2019, Nature Medicine.

[51]  Thomas E. Hughes,et al.  Safe and efficient peripheral blood stem cell collection in patients with sickle cell disease using plerixafor , 2019, Haematologica.

[52]  R. Morgan,et al.  Improved Titer and Gene Transfer by Lentiviral Vectors Using Novel, Small β-Globin Locus Control Region Elements. , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[53]  H. Deeg,et al.  Impact of Conditioning Intensity of Allogeneic Transplantation for Acute Myeloid Leukemia With Genomic Evidence of Residual Disease. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[54]  A. Chaudhury,et al.  Single-cell modeling of routine clinical blood tests reveals transient dynamics of human response to blood loss , 2019, eLife.

[55]  R. D. Hawkins,et al.  Targeted Integration and High-Level Transgene Expression in AAVS1 Transgenic Mice after In Vivo HSC Transduction with HDAd5/35++ Vectors. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[56]  D. K. Wood,et al.  High-throughput assessment of hemoglobin polymer in single red blood cells from sickle cell patients under controlled oxygen tension , 2019, Proceedings of the National Academy of Sciences.

[57]  M. DeBaun,et al.  Are genetic approaches still needed to cure sickle cell disease? , 2019, The Journal of clinical investigation.

[58]  D. K. Wood,et al.  A microfluidic platform for simultaneous quantification of oxygen-dependent viscosity and shear thinning in sickle cell blood , 2019, APL bioengineering.

[59]  J. Tisdale,et al.  Resolution of Sickle Cell Disease Manifestations in Patients Treated with Lentiglobin Gene Therapy: Updated Results from the Phase 1/2 Hgb-206 Group C Study , 2019, Blood.

[60]  Anthony E. Boitano,et al.  A Single Dose of CD117 Antibody Drug Conjugate Enables Autologous Gene-Modified Hematopoietic Stem Cell Transplant (Gene Therapy) in Nonhuman Primates , 2019, Blood.

[61]  I. Weissman,et al.  Non-Genotoxic Anti-CD117 Antibody Conditioning Results in Successful Hematopoietic Stem Cell Engraftment in Patients with Severe Combined Immunodeficiency , 2019, Blood.

[62]  I. Weissman,et al.  An All Antibody Approach for Conditioning Bone Marrow for Hematopoietic Stem Cell Transplantation with Anti-cKIT and Anti-CD47 in Non-Human Primates , 2019, Blood.

[63]  David A. Williams,et al.  Preliminary Results of a Phase 1/2 Clinical Study of Zinc Finger Nuclease-Mediated Editing of BCL11A in Autologous Hematopoietic Stem Cells for Transfusion-Dependent Beta Thalassemia , 2019, Blood.

[64]  Shondra M. Pruett-Miller,et al.  Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. , 2019, Blood advances.

[65]  Junjiu Huang,et al.  Off-target effects of cytidine base editor and adenine base editor: What can we do? , 2019, Journal of genetics and genomics = Yi chuan xue bao.

[66]  J. Kaiser,et al.  Gates and NIH join forces on HIV and sickle cell diseases. , 2019, Science.

[67]  J. Wagner,et al.  Effect of donor type and conditioning regimen intensity on allogeneic transplantation outcomes in patients with sickle cell disease: a retrospective multicentre, cohort study. , 2019, The Lancet. Haematology.

[68]  David R. Liu,et al.  Search-and-replace genome editing without double-strand breaks or donor DNA , 2019, Nature.

[69]  D. Boger,et al.  Resveratrol trimer enhances gene delivery to hematopoietic stem cells by reducing antiviral restriction at endosomes. , 2019, Blood.

[70]  A. Miccio,et al.  Lentiviral and genome-editing strategies for the treatment of β-hemoglobinopathies. , 2019, Blood.

[71]  S. Subramaniam,et al.  Nongenotoxic antibody-drug conjugate conditioning enables safe and effective platelet gene therapy of hemophilia A mice. , 2019, Blood advances.

[72]  B. Williams,et al.  Antibody Therapies for Acute Myeloid Leukemia: Unconjugated, Toxin-Conjugated, Radio-Conjugated and Multivalent Formats , 2019, Journal of clinical medicine.

[73]  Joseph D. Long,et al.  Editing the Sickle Cell Disease Mutation in Human Hematopoietic Stem Cells: Comparison of Endonucleases and Homologous Donor Templates. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[74]  A. Scharenberg,et al.  Therapeutically relevant engraftment of a CRISPR-Cas9–edited HSC-enriched population with HbF reactivation in nonhuman primates , 2019, Science Translational Medicine.

[75]  J. Tisdale,et al.  Bone marrow characterization in sickle cell disease: inflammation and stress erythropoiesis lead to suboptimal CD34 recovery , 2019, British journal of haematology.

[76]  Daesik Kim,et al.  Evaluating and Enhancing Target Specificity of Gene-Editing Nucleases and Deaminases. , 2019, Annual review of biochemistry.

[77]  Hui Yang,et al.  Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis , 2019, Nature.

[78]  A. Schambach,et al.  Enhancing Lentiviral and Alpharetroviral Transduction of Human Hematopoietic Stem Cells for Clinical Application , 2019, Molecular therapy. Methods & clinical development.

[79]  A. Scharenberg,et al.  In Vivo Outcome of Homology-Directed Repair at the HBB Gene in HSC Using Alternative Donor Template Delivery Methods , 2019, Molecular therapy. Nucleic acids.

[80]  Chuanfeng Wu,et al.  Aberrant Clonal Hematopoiesis following Lentiviral Vector Transduction of HSPCs in a Rhesus Macaque. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[81]  M. DeBaun,et al.  Haploidentical Bone Marrow Transplantation with Post-Transplantation Cyclophosphamide Plus Thiotepa Improves Donor Engraftment in Patients with Sickle Cell Anemia: Results of an International Learning Collaborative. , 2019, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[82]  M. Porteus,et al.  Highly efficient editing of the β-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease , 2019, Nucleic acids research.

[83]  Adrian Pickar-Oliver,et al.  The next generation of CRISPR–Cas technologies and applications , 2019, Nature Reviews Molecular Cell Biology.

[84]  Martin J. Aryee,et al.  Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors , 2019, Nature.

[85]  D. Kohn,et al.  PGE2 and Poloxamer Synperonic F108 Enhance Transduction of Human HSPCs with a β-Globin Lentiviral Vector , 2019, Molecular therapy. Methods & clinical development.

[86]  C. Huff,et al.  Effect of increased dose of total body irradiation on graft failure associated with HLA-haploidentical transplantation in patients with severe haemoglobinopathies: a prospective clinical trial. , 2019, The Lancet. Haematology.

[87]  Chunyan Ren,et al.  Highly efficient therapeutic gene editing of human hematopoietic stem cells , 2019, Nature Medicine.

[88]  Daesik Kim,et al.  Genome-wide target specificity of CRISPR RNA-guided adenine base editors , 2019, Nature Biotechnology.

[89]  I. Weissman,et al.  Toxicity-Free Hematopoietic Stem Cell Engraftment Achieved with Anti-CD117 Monoclonal Antibody Conditioning , 2019, Biology of Blood and Marrow Transplantation.

[90]  Derrick J. Rossi,et al.  Hematopoietic chimerism and donor-specific skin allograft tolerance after non-genotoxic CD117 antibody-drug-conjugate conditioning in MHC-mismatched allotransplantation , 2019, Nature Communications.

[91]  D. Scadden,et al.  Selective hematopoietic stem cell ablation using CD117-antibody-drug-conjugates enables safe and effective transplantation with immunity preservation , 2019, Nature Communications.

[92]  S. Orkin,et al.  Emerging Genetic Therapy for Sickle Cell Disease. , 2019, Annual review of medicine.

[93]  L. Naldini Genetic engineering of hematopoiesis: current stage of clinical translation and future perspectives , 2019, EMBO molecular medicine.

[94]  S. Perrotta,et al.  Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent ß-thalassemia , 2019, Nature Medicine.

[95]  Junjiu Huang,et al.  Genome-wide profiling of adenine base editor specificity by EndoV-seq , 2019, Nature Communications.

[96]  P. Mangeot,et al.  Genome editing in primary cells and in vivo using viral-derived Nanoblades loaded with Cas9-sgRNA ribonucleoproteins , 2019, Nature Communications.

[97]  M. Porteus,et al.  Optimization of CRISPR/Cas9 Delivery to Human Hematopoietic Stem and Progenitor Cells for Therapeutic Genomic Rearrangements. , 2019, Molecular therapy : the journal of the American Society of Gene Therapy.

[98]  Z. Izsvák,et al.  In vivo hematopoietic stem cell gene therapy ameliorates murine thalassemia intermedia , 2018, The Journal of clinical investigation.

[99]  D. Kohn Gene therapy for blood diseases. , 2019, Current opinion in biotechnology.

[100]  S. Wyman,et al.  Timed inhibition of CDC7 increases CRISPR-Cas9 mediated templated repair , 2018, bioRxiv.

[101]  K. Quinlan,et al.  Wake-up Sleepy Gene: Reactivating Fetal Globin for β-Hemoglobinopathies. , 2018, Trends in genetics : TIG.

[102]  S. Tsai,et al.  Illuminating the genome-wide activity of genome editors for safe and effective therapeutics , 2018, Genome Biology.

[103]  Weiliang Shi,et al.  Outcomes for Initial Patient Cohorts with up to 33 Months of Follow-up in the Hgb-206 Phase 1 Trial , 2018, Blood.

[104]  F. Bushman,et al.  Gene Therapy for Sickle Cell Anemia Using a Modified Gamma Globin Lentivirus Vector and Reduced Intensity Conditioning Transplant Shows Promising Correction of the Disease Phenotype , 2018, Blood.

[105]  Weiliang Shi,et al.  Current Results of Lentiglobin Gene Therapy in Patients with Severe Sickle Cell Disease Treated Under a Refined Protocol in the Phase 1 Hgb-206 Study , 2018, Blood.

[106]  A. Schambach,et al.  Pre-clinical Development of a Lentiviral Vector Expressing the Anti-sickling βAS3 Globin for Gene Therapy for Sickle Cell Disease , 2018, Molecular therapy. Methods & clinical development.

[107]  David A. Williams,et al.  Successful hematopoietic stem cell mobilization and apheresis collection using plerixafor alone in sickle cell patients. , 2018, Blood advances.

[108]  L. Biasco,et al.  Dynamics of genetically engineered hematopoietic stem and progenitor cells after autologous transplantation in humans , 2018, Nature Medicine.

[109]  Ahmad Sabry Mohamad,et al.  Human hemoglobin G-Makassar variant masquerading as sickle cell anemia , 2018, Hematology reports.

[110]  R. Medema,et al.  A limited number of double-strand DNA breaks is sufficient to delay cell cycle progression , 2018, bioRxiv.

[111]  A. Bradley,et al.  Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements , 2018, Nature Biotechnology.

[112]  Jussi Taipale,et al.  CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response , 2018, Nature Medicine.

[113]  Gregory McAllister,et al.  p53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells , 2018, Nature Medicine.

[114]  R. Bak,et al.  Priming Human Repopulating Hematopoietic Stem and Progenitor Cells for Cas9/sgRNA Gene Targeting , 2018, Molecular therapy. Nucleic acids.

[115]  C. von Kalle,et al.  Gene Therapy in Patients with Transfusion‐Dependent β‐Thalassemia , 2018, The New England journal of medicine.

[116]  Martha L. Bulyk,et al.  Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch , 2018, Cell.

[117]  Mildred K Cho,et al.  Beyond Consent: Building Trusting Relationships With Diverse Populations in Precision Medicine Research , 2018, The American journal of bioethics : AJOB.

[118]  V. Bonham,et al.  The Role of the Health Care Provider in Building Trust Between Patients and Precision Medicine Research Programs , 2018, The American journal of bioethics : AJOB.

[119]  Morgan L. Maeder,et al.  UDiTaS™, a genome editing detection method for indels and genome rearrangements , 2018, BMC Genomics.

[120]  D. Weatherall,et al.  Sickle cell disease , 2018, Nature Reviews Disease Primers.

[121]  Laura J. Norton,et al.  Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding , 2018, Nature Genetics.

[122]  A. Miccio,et al.  Plerixafor enables safe, rapid, efficient mobilization of hematopoietic stem cells in sickle cell disease patients after exchange transfusion , 2018, Haematologica.

[123]  M. Sadelain,et al.  Safety and efficacy of plerixafor dose escalation for the mobilization of CD34+ hematopoietic progenitor cells in patients with sickle cell disease: interim results , 2018, Haematologica.

[124]  Alireza Paikari,et al.  Fetal haemoglobin induction in sickle cell disease , 2018, British journal of haematology.

[125]  L. Krishnamurti,et al.  Experiences and Decision Making in Hematopoietic Stem Cell Transplant in Sickle Cell Disease: Patients' and Caregivers' Perspectives. , 2017, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[126]  E. Vichinsky,et al.  Variability of homozygous sickle cell disease: The role of alpha and beta globin chain variation and other factors. , 2017, Blood cells, molecules & diseases.

[127]  P. Kharchenko,et al.  Rapid Mobilization Reveals a Highly Engraftable Hematopoietic Stem Cell , 2016, Cell.

[128]  M. Walters,et al.  The case for HLA-identical sibling hematopoietic stem cell transplantation in children with symptomatic sickle cell anemia. , 2017, Blood advances.

[129]  E. Clayton,et al.  Primum non nocere: the case against transplant for children with sickle cell anemia without progressive end-organ disease. , 2017, Blood advances.

[130]  D. Calvet,et al.  Low fetal hemoglobin percentage is associated with silent brain lesions in adults with homozygous sickle cell disease. , 2017, Blood advances.

[131]  M. Steinberg,et al.  Fetal hemoglobin in sickle cell anemia: The Arab‐Indian haplotype and new therapeutic agents , 2017, American journal of hematology.

[132]  J. Tisdale,et al.  At least 20% donor myeloid chimerism is necessary to reverse the sickle phenotype after allogeneic HSCT. , 2017, Blood.

[133]  H. Bang,et al.  Increased risk of leukemia among sickle cell disease patients in California. , 2017, Blood.

[134]  D. Kohn,et al.  Improving Gene Therapy Efficiency through the Enrichment of Human Hematopoietic Stem Cells. , 2017, Molecular therapy : the journal of the American Society of Gene Therapy.

[135]  J. Tisdale,et al.  Bone Marrow as a Hematopoietic Stem Cell Source for Gene Therapy in Sickle Cell Disease: Evidence from Rhesus and SCD Patients. , 2017, Human gene therapy. Clinical development.

[136]  D. Jacobsohn,et al.  Unrelated Umbilical Cord Blood Transplantation for Sickle Cell Disease Following Reduced-Intensity Conditioning: Results of a Phase I Trial. , 2017, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[137]  Megan E. McNerney,et al.  Therapy-related myeloid neoplasms: when genetics and environment collide , 2017, Nature Reviews Cancer.

[138]  A. Nagler,et al.  Single Dose of the CXCR4 Antagonist BL-8040 Induces Rapid Mobilization for the Collection of Human CD34+ Cells in Healthy Volunteers , 2017, Clinical Cancer Research.

[139]  G. Loewenstein,et al.  Proponent or collaborative: Physician perspectives and approaches to disease modifying therapies in sickle cell disease , 2017, PloS one.

[140]  P. Malik,et al.  Patient Perspectives on Gene Transfer Therapy for Sickle Cell Disease , 2017, Advances in Therapy.

[141]  W. Eaton,et al.  Treating sickle cell disease by targeting HbS polymerization. , 2017, Blood.

[142]  J. Powell,et al.  Cyclophosphamide improves engraftment in patients with SCD and severe organ damage who undergo haploidentical PBSCT. , 2017, Blood advances.

[143]  Daesik Kim,et al.  Genome-wide target specificities of CRISPR RNA-guided programmable deaminases , 2017, Nature Biotechnology.

[144]  J. Kurtzberg,et al.  Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. , 2017, Blood.

[145]  X. Yang,et al.  Long-Term Engraftment and Fetal Globin Induction upon BCL11A Gene Editing in Bone-Marrow-Derived CD34+ Hematopoietic Stem and Progenitor Cells , 2017, Molecular therapy. Methods & clinical development.

[146]  J. Davies,et al.  Gene Therapy in a Patient with Sickle Cell Disease. , 2017, The New England journal of medicine.

[147]  S. Nilsson,et al.  New agents in HSC mobilization , 2017, International Journal of Hematology.

[148]  A. Wach,et al.  Mobilization of hematopoietic stem cells with the novel CXCR4 antagonist POL6326 (balixafortide) in healthy volunteers—results of a dose escalation trial , 2017, Journal of Translational Medicine.

[149]  J. Tisdale,et al.  Interim Results from a Phase 1/2 Clinical Study of Lentiglobin Gene Therapy for Severe Sickle Cell Disease , 2016 .

[150]  Sruthi Mantri,et al.  CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells , 2016, Nature.

[151]  Dana Carroll,et al.  Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells , 2016, Science Translational Medicine.

[152]  Matthew C. Canver,et al.  Lineage-specific BCL11A knockdown circumvents toxicities and reverses sickle phenotype. , 2016, The Journal of clinical investigation.

[153]  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.

[154]  I. Weissman,et al.  Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy , 2016, Science Translational Medicine.

[155]  David A. Williams,et al.  Mathematical modeling of erythrocyte chimerism informs genetic intervention strategies for sickle cell disease , 2016, American journal of hematology/oncology.

[156]  Chavis A. Patterson,et al.  Mistrust of Pediatric Sickle Cell Disease Clinical Trials Research. , 2016, American journal of preventive medicine.

[157]  D. Scadden,et al.  Non-genotoxic conditioning for hematopoietic stem cell transplantation using a hematopoietic-cell-specific internalizing immunotoxin , 2016, Nature Biotechnology.

[158]  G. Lettre,et al.  Fetal haemoglobin in sickle-cell disease: from genetic epidemiology to new therapeutic strategies , 2016, The Lancet.

[159]  A. Raj,et al.  Enhancer Regulation of Transcriptional Bursting Parameters Revealed by Forced Chromatin Looping. , 2016, Molecular cell.

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

[161]  A. Baruchel,et al.  Clinical and haematological risk factors for cerebral macrovasculopathy in a sickle cell disease newborn cohort: a prospective study , 2016, British journal of haematology.

[162]  Matthew C. Canver,et al.  Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin , 2016, Science.

[163]  P. Nederkoorn,et al.  Risk factor analysis of cerebral white matter hyperintensities in children with sickle cell disease , 2016, British journal of haematology.

[164]  Matthew C. Canver,et al.  miRNA-embedded shRNAs for Lineage-specific BCL11A Knockdown and Hemoglobin F Induction. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

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

[166]  B. Sandmaier,et al.  (211)Astatine-Conjugated Monoclonal CD45 Antibody-Based Nonmyeloablative Conditioning for Stem Cell Gene Therapy. , 2015, Human gene therapy.

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

[168]  Christopher A. Miller,et al.  The Role of TP53 Mutations in the Origin and Evolution of Therapy-Related AML , 2014, Nature.

[169]  J. Herrick,et al.  Peculiar elongated and sickle-shaped red blood corpuscles in a case of severe anemia. , 2014, JAMA.

[170]  William J Savage,et al.  Management of sickle cell disease: summary of the 2014 evidence-based report by expert panel members. , 2014, JAMA.

[171]  M. Sorror,et al.  Radiolabeled anti-CD45 antibody with reduced-intensity conditioning and allogeneic transplantation for younger patients with advanced acute myeloid leukemia or myelodysplastic syndrome. , 2014, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[172]  Paola Sebastiani,et al.  Fetal hemoglobin in sickle cell anemia: a glass half full? , 2013, Blood.

[173]  A. Nagler,et al.  The High-Affinity CXCR4 Antagonist BKT140 Is Safe and Induces a Robust Mobilization of Human CD34+ Cells in Patients with Multiple Myeloma , 2013, Clinical Cancer Research.

[174]  J. Jurcic Radioimmunotherapy for hematopoietic cell transplantation. , 2013, Immunotherapy.

[175]  P. Sebastiani,et al.  Genetic modifiers of sickle cell disease , 2012, American journal of hematology.

[176]  J. Miller,et al.  Associated risk factors for silent cerebral infarcts in sickle cell anemia: low baseline hemoglobin, sex, and relative high systolic blood pressure. , 2012, Blood.

[177]  B. Sandmaier,et al.  Conditioning with α-emitter based radioimmunotherapy in canine allogeneic hematopoietic cell transplantation , 2012, Chimerism.

[178]  I. Bhan,et al.  Fetal haemoglobin levels and haematological characteristics of compound heterozygotes for haemoglobin S and deletional hereditary persistence of fetal haemoglobin , 2012, British journal of haematology.

[179]  Debashis Sahoo,et al.  Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age , 2011, Proceedings of the National Academy of Sciences.

[180]  P. Tazzari,et al.  Quantitatively different red cell/nucleated cell chimerism in patients with long-term, persistent hematopoietic mixed chimerism after bone marrow transplantation for thalassemia major or sickle cell disease , 2010, Haematologica.

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

[182]  J. Haldane,et al.  THE RATE OF MUTATION OF HUMAN GENES , 2010 .

[183]  B. Andersson,et al.  Busulfan in hematopoietic stem cell transplantation. , 2009, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[184]  J. Tisdale,et al.  Granulocyte colony-stimulating factor (G-CSF) administration in individuals with sickle cell disease: time for a moratorium? , 2009, Cytotherapy.

[185]  F. Grosveld,et al.  The beta-globin nuclear compartment in development and erythroid differentiation. , 2003, Nature genetics.

[186]  Wouter de Laat,et al.  The β-globin nuclear compartment in development and erythroid differentiation , 2003, Nature Genetics.

[187]  J. Burke,et al.  Cytoreduction with iodine-131-anti-CD33 antibodies before bone marrow transplantation for advanced myeloid leukemias , 2003, Bone Marrow Transplantation.

[188]  B. Rerkamnuaychoke,et al.  Hb G Makassar (beta 6:Glu-Ala) in a Thai family. , 2002, Journal of the Medical Association of Thailand = Chotmaihet thangphaet.

[189]  V. Viprakasit,et al.  Hb G-MAKASSAR [β6(A3)Glu→Ala; CODON 6 (G A G→G C G)]: MOLECULAR CHARACTERIZATION, CLINICAL, AND HEMATOLOGICAL EFFECTS , 2002, Hemoglobin.

[190]  K. Sullivan,et al.  Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia. , 2001, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[191]  Esther Nzewi,et al.  Malevolent ogbanje: recurrent reincarnation or sickle cell disease? , 2001, Social science & medicine.

[192]  Other pharmaceutical agents. , 2000, IARC monographs on the evaluation of carcinogenic risks to humans.

[193]  I. Bernstein,et al.  Phase I study of (131)I-anti-CD45 antibody plus cyclophosphamide and total body irradiation for advanced acute leukemia and myelodysplastic syndrome. , 1999, Blood.

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

[195]  Scott T. Miller,et al.  Cerebrovascular accidents in sickle cell disease: rates and risk factors. , 1998, Blood.

[196]  P. Lane Sickle cell disease. , 1996, Pediatric clinics of North America.

[197]  D. Scheinberg,et al.  A phase I trial of monoclonal antibody M195 in acute myelogenous leukemia: specific bone marrow targeting and internalization of radionuclide. , 1991, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[198]  A. Bank,et al.  Gene Transfer , 1989 .

[199]  J. Onwubalili SICKLE-CELL ANAEMIA: AN EXPLANATION FOR THE ANCIENT MYTH OF REINCARNATION IN NIGERIA , 1983, The Lancet.

[200]  Y. Kan,et al.  ANTENATAL DIAGNOSIS OF SICKLE-CELL ANÆMIA BY D.N.A. ANALYSIS OF AMNIOTIC-FLUID CELLS , 1978, The Lancet.

[201]  S. Oemijati,et al.  Hemoglobin G Makassar: β6 Glu→Ala , 1970 .

[202]  S. Oemijati,et al.  Hemoglobin G Makassar: beta-6 Glu leads to Ala. , 1970, Biochimica et biophysica acta.

[203]  V. Ingram,et al.  A Specific Chemical Difference Between the Globins of Normal Human and Sickle-Cell Anæmia Hæmoglobin , 1956, Nature.

[204]  A. Allison,et al.  Protection Afforded by Sickle-cell Trait Against Subtertian Malarial Infection , 1954, British medical journal.

[205]  L. Pauling,et al.  Sickle cell anemia a molecular disease. , 1949, Science.

[206]  J. Neel The Inheritance of Sickle Cell Anemia. , 1949, Science.

[207]  E. A. Beet The genetics of the sickle-cell trait in a Bantu tribe. , 1949, Annals of eugenics.