Gene Editing and Genotoxicity: Targeting the Off-Targets
暂无分享,去创建一个
Georges Blattner | Alessia Cavazza | Adrian J. Thrasher | Giandomenico Turchiano | A. Thrasher | G. Turchiano | A. Cavazza | Georges Blattner | Alessia Cavazza
[1] B. van Steensel,et al. Easy quantitative assessment of genome editing by sequence trace decomposition , 2014, Nucleic acids research.
[2] Peng Qiu,et al. COSMID: A Web-based Tool for Identifying and Validating CRISPR/Cas Off-target Sites , 2014, Molecular therapy. Nucleic acids.
[3] Daesik Kim,et al. Evaluating and Enhancing Target Specificity of Gene-Editing Nucleases and Deaminases. , 2019, Annual review of biochemistry.
[4] BiascoLuca. Integration Site Analysis in Gene Therapy Patients: Expectations and Reality. , 2017 .
[5] Qiaobing Xu,et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents , 2017, Nature.
[6] 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.
[7] Teresa M. Przytycka,et al. DNA Break Mapping Reveals Topoisomerase II Activity Genome-Wide , 2014, International journal of molecular sciences.
[8] Wei Chen,et al. Retargeting sleeping beauty transposon insertions by engineered zinc finger DNA-binding domains. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.
[9] Mazhar Adli,et al. Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease , 2014, Nature Biotechnology.
[10] Leslie S. Edwards,et al. Mapping the genomic landscape of CRISPR–Cas9 cleavage , 2017, Nature Methods.
[11] Martin J. Aryee,et al. GUIDE-Seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases , 2014, Nature Biotechnology.
[12] J. Keith Joung,et al. High frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells , 2013, Nature Biotechnology.
[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] Adam Bagg,et al. Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. , 2003, Molecular genetics and metabolism.
[15] Elif Karaca,et al. Simple and rapid in vivo generation of chromosomal rearrangements using CRISPR/Cas9 technology. , 2014, Cell reports.
[16] J. Sklar,et al. Detection of mutations by cleavage of DNA heteroduplexes with bacteriophage resolvases , 1995, Nature Genetics.
[17] Jesse R. Dixon,et al. Topological Domains in Mammalian Genomes Identified by Analysis of Chromatin Interactions , 2012, Nature.
[18] David R. Liu,et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage , 2016, Nature.
[19] Christof von Kalle,et al. and insertional genotoxicity Cell culture assays reveal the importance of retroviral vector design for , 2006 .
[20] A. Nussenzweig,et al. DNA Breaks and End Resection Measured Genome-wide by End Sequencing. , 2016, Molecular cell.
[21] Andreas Gogol-Döring,et al. RNA-guided retargeting of Sleeping Beauty transposition in human cells , 2020, eLife.
[22] Jérôme Larghero,et al. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia , 2010, Nature.
[23] V. Myer,et al. Characterization of the interplay between DNA repair and CRISPR/Cas9-induced DNA lesions at an endogenous locus , 2017, Nature Communications.
[24] Georges Blattner,et al. Targeted gene correction of human hematopoietic stem cells for the treatment of Wiskott - Aldrich Syndrome , 2020, Nature Communications.
[25] 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.
[26] J. Durocher,et al. Mutation detection using Surveyor nuclease. , 2004, BioTechniques.
[27] Charles D. Yeh,et al. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq , 2018, Science.
[28] Luca Biasco,et al. Lentiviral gene therapy for X-linked chronic granulomatous disease , 2020, Nature Medicine.
[29] Israel Steinfeld,et al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells , 2015, Nature Biotechnology.
[30] Nicole M. Gaudelli,et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage , 2017, Nature.
[31] J. Keith Joung,et al. CRISPR C-to-G base editors for inducing targeted DNA transversions in human cells , 2020, Nature Biotechnology.
[32] Daesik Kim,et al. Genome-wide target specificity of CRISPR RNA-guided adenine base editors , 2019, Nature Biotechnology.
[33] Christopher Baum,et al. A modified γ-retrovirus vector for X-linked severe combined immunodeficiency. , 2014, The New England journal of medicine.
[34] Ivan Merelli,et al. Precise Gene Editing Preserves Hematopoietic Stem Cell Function following Transient p53-Mediated DNA Damage Response , 2019, Cell stem cell.
[35] Bruce R. Conklin,et al. CRISPR off-target detection with DISCOVER-seq , 2020, Nature Protocols.
[36] Zeger Debyser,et al. Towards a Safer, More Randomized Lentiviral Vector Integration Profile Exploring Artificial LEDGF Chimeras , 2016, PloS one.
[37] Pratima Chowdary,et al. Adenovirus-associated virus vector-mediated gene transfer in hemophilia B. , 2011, The New England journal of medicine.
[38] Jeffrey C. Miller,et al. An unbiased genome-wide analysis of zinc-finger nuclease specificity , 2011, Nature Biotechnology.
[39] Jin-Soo Kim,et al. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases , 2014, Bioinform..
[40] A. Zanghellini,et al. A novel engineered meganuclease induces homologous recombination in yeast and mammalian cells. , 2003, Nucleic acids research.
[41] Jennifer A. Doudna,et al. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity , 2018, Science.
[42] Luca Biasco. Integration Site Analysis in Gene Therapy Patients: Expectations and Reality. , 2017, Human gene therapy.
[43] Maximilian Haeussler,et al. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens , 2018, Nucleic Acids Res..
[44] Imen Lassadi,et al. High‐resolution profiling of γH2AX around DNA double strand breaks in the mammalian genome , 2010, The EMBO journal.
[45] Hang Wu,et al. Nick-seq for single-nucleotide resolution genomic maps of DNA modifications and damage , 2019, bioRxiv.
[46] David R. Liu,et al. Search-and-replace genome editing without double-strand breaks or donor DNA , 2019, Nature.
[47] K. Cornetta,et al. Gene transfer into humans--immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. , 1990, The New England journal of medicine.
[48] G. Cullot,et al. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations , 2019, Nature Communications.
[49] Kevin T. Zhao,et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity , 2017, Science Advances.
[50] Jin-Soo Kim,et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases , 2014, Genome research.
[51] Morgan L. Maeder,et al. UDiTaS™, a genome editing detection method for indels and genome rearrangements , 2018, BMC Genomics.
[52] Barry L. Stoddard,et al. The homing endonuclease I-CreI uses three metals, one of which is shared between the two active sites , 2001, Nature Structural Biology.
[53] Chunyan Ren,et al. Therapeutic base editing of human hematopoietic stem cells , 2020, Nature Medicine.
[54] Daesik Kim,et al. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins , 2014, Genome research.
[55] 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.
[56] Christine Kinnon,et al. Mutations in TNFRSF13B Encoding TACI Are Associated With Common Variable Immunodeficiency in Humans , 2006, Pediatrics.
[57] Gang Bao,et al. A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human haematopoietic stem and progenitor cells , 2018, Nature Medicine.
[58] Howard Y. Chang,et al. CRISPR-engineered T cells in patients with refractory cancer , 2020, Science.
[59] Cameron S. Osborne,et al. LMO2-Associated Clonal T Cell Proliferation in Two Patients after Gene Therapy for SCID-X1 , 2003, Science.
[60] Richard L. Frock,et al. Detecting DNA double-stranded breaks in mammalian genomes by linear amplification–mediated high-throughput genome-wide translocation sequencing , 2016, Nature Protocols.
[61] Jong-il Kim,et al. Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-target effects in human cells , 2015, Nature Methods.
[62] Yilong Li,et al. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library , 2013, Nature Biotechnology.
[63] Gang Wang,et al. Targeted and genome-wide sequencing reveal single nucleotide variations impacting specificity of Cas9 in human stem cells , 2014, Nature Communications.
[64] Andreas Reinisch,et al. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6 , 2017, eLife.
[65] Nicholas A. Rossi,et al. Inference of CRISPR Edits from Sanger Trace Data , 2019, bioRxiv.
[66] W. F. Anderson,et al. September 14, 1990: the beginning. , 1990, Human gene therapy.
[67] Daesik Kim,et al. Genome-wide target specificities of CRISPR RNA-guided programmable deaminases , 2017, Nature Biotechnology.
[68] 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.
[69] Qiu-Xiang Cheng,et al. CRISPR-Cas12a has both cis- and trans-cleavage activities on single-stranded DNA , 2018, Cell Research.
[70] Michel C. Nussenzweig,et al. Translocation-Capture Sequencing Reveals the Extent and Nature of Chromosomal Rearrangements in B Lymphocytes , 2011, Cell.
[71] A. Bogdanove,et al. TAL Effectors: Customizable Proteins for DNA Targeting , 2011, Science.
[72] Daesik Kim,et al. DIG-seq: a genome-wide CRISPR off-target profiling method using chromatin DNA , 2018, Genome research.
[73] Zeger Debyser,et al. LEDGF hybrids efficiently retarget lentiviral integration into heterochromatin. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.
[74] Jeffrey C. Miller,et al. Highly efficient endogenous human gene correction using designed zinc-finger nucleases , 2005, Nature.
[75] Yuan Ping,et al. Off-Targeting of Base Editors: BE3 but not ABE induces substantial off-target single nucleotide variants , 2019, Signal Transduction and Targeted Therapy.
[76] Giuseppe Petrosino,et al. Genome-wide Nucleotide-Resolution Mapping of DNA Replication Patterns, Single-Strand Breaks, and Lesions by GLOE-Seq , 2020, Molecular cell.
[77] Melanie Boerries,et al. Quantitative Evaluation of Chromosomal Rearrangements in Primary Gene-Edited Human Stem Cells by Preclinical CAST-Seq , 2020, SSRN Electronic Journal.
[78] Jin-Soo Kim,et al. Measuring and Reducing Off-Target Activities of Programmable Nucleases Including CRISPR-Cas9 , 2015, Molecules and cells.
[79] David R. Liu,et al. Analysis and minimization of cellular RNA editing by DNA adenine base editors , 2019, Science Advances.
[80] Philippe Horvath,et al. The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli , 2011, Nucleic acids research.
[81] Cheng-Zhong Zhang,et al. Chromothripsis as an on-target consequence of CRISPR-Cas9 genome editing , 2020, Nature Genetics.
[82] Hang Wu,et al. Nick-seq for single-nucleotide resolution genomic maps of DNA modifications and damage , 2020, Nucleic acids research.
[83] David A. Scott,et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells , 2014, Nature Biotechnology.
[84] Dan Liu,et al. Genome-wide profiling of adenine base editor specificity by EndoV-seq , 2019, Nature Communications.
[85] Giacomo Cavalli,et al. Organization and function of the 3 D genome , 2022 .
[86] Jenny A. Greig,et al. ITR-Seq, a next-generation sequencing assay, identifies genome-wide DNA editing sites in vivo following adeno-associated viral vector-mediated genome editing , 2020, BMC Genomics.
[87] David A. Scott,et al. In vivo genome editing using Staphylococcus aureus Cas9 , 2015, Nature.
[88] Philipp Kapranov,et al. Novel approach reveals genomic landscapes of single-strand DNA breaks with nucleotide resolution in human cells , 2019, Nature Communications.
[89] Feng Zhang,et al. Breaks Labeling in situ and sequencing (BLISS) , 2017 .
[90] David M Weinstock,et al. Induction of chromosomal translocations in mouse and human cells using site-specific endonucleases. , 2008, Journal of the National Cancer Institute. Monographs.
[91] Samuel H. Sternberg,et al. Transposon-encoded CRISPR–Cas systems direct RNA-guided DNA integration , 2019, Nature.
[92] Ruochi Zhang,et al. CHANGE-seq reveals genetic and epigenetic effects on CRISPR-Cas9 genome-wide activity , 2020, Nature Biotechnology.
[93] Luca Biasco,et al. Lentiviral Hematopoietic Stem Cell Gene Therapy in Patients with Wiskott-Aldrich Syndrome , 2013, Science.
[94] Jacqueline Corrigan-Curay,et al. Genome Editing Technologies: Defining a Path to Clinic: Genomic Editing: Establishing Preclinical Toxicology Standards, Bethesda, Maryland 10 June 2014. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.
[95] Alessandro Aiuti,et al. Hot spots of retroviral integration in human CD34+ hematopoietic cells. , 2007, Blood.
[96] Richard L. Frock,et al. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases , 2014, Nature Biotechnology.
[97] Giacomo Cavalli,et al. Organization and function of the 3D genome , 2016, Nature Reviews Genetics.
[98] Toni Cathomen,et al. Genotoxicity in gene therapy: an account of vector integration and designer nucleases. , 2008, Current opinion in molecular therapeutics.
[99] 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.
[100] C. Dunbar,et al. The MDS1-EVI1 gene complex as a retrovirus integration site: impact on behavior of hematopoietic cells and implications for gene therapy. , 2008, Molecular therapy : the journal of the American Society of Gene Therapy.
[101] T. Rabbitts,et al. Activation of the T-cell oncogene LMO2 after gene therapy for X-linked severe combined immunodeficiency. , 2004, The New England journal of medicine.
[102] Daniel Capurso,et al. DNA Repair Profiling Reve als Nonrandom Outcomes at Cas 9-Mediated Breaks Graphical Abstract Highlights , 2016 .
[103] Alessandro Romanel,et al. A highly specific SpCas9 variant is identified by in vivo screening in yeast , 2018, Nature Biotechnology.
[104] Martin Kircher,et al. Double indexing overcomes inaccuracies in multiplex sequencing on the Illumina platform , 2011, Nucleic acids research.
[105] F. Gage,et al. Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. , 1996, Proceedings of the National Academy of Sciences of the United States of America.
[106] George M. Church,et al. CHOPCHOP: a CRISPR/Cas9 and TALEN web tool for genome editing , 2014, Nucleic Acids Res..
[107] M. Rowicka,et al. Nucleotide-resolution DNA double-strand breaks mapping by next-generation sequencing , 2013, Nature Methods.
[108] Yong-Sam Kim,et al. Unbiased investigation of specificities of prime editing systems in human cells , 2020, Nucleic acids research.
[109] Erik L. G. Wernersson,et al. BLISS is a versatile and quantitative method for genome-wide profiling of DNA double-strand breaks , 2017, Nature Communications.
[110] Anob M. Chakrabarti,et al. Target-Specific Precision of CRISPR-Mediated Genome Editing , 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] J. Keith Joung,et al. 731. High-Fidelity CRISPR-Cas9 Nucleases with No Detectable Genome-Wide Off-Target Effects , 2016 .
[113] Stefan Posch,et al. TALENoffer: genome-wide TALEN off-target prediction , 2013, Bioinform..
[114] J. Doudna,et al. A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.
[115] Jon P. Connelly,et al. CRIS.py: A Versatile and High-throughput Analysis Program for CRISPR-based Genome Editing , 2019, Scientific Reports.
[116] Adam James Waite,et al. An improved zinc-finger nuclease architecture for highly specific genome editing , 2007, Nature Biotechnology.
[117] Jin-Soo Kim,et al. Genome-wide target specificities of CRISPR-Cas9 nucleases revealed by multiplex Digenome-seq , 2016, Genome research.
[118] Christine Kinnon,et al. Gammaretrovirus-mediated correction of SCID-X1 is associated with skewed vector integration site distribution in vivo. , 2007, The Journal of clinical investigation.
[119] A. Iafrate,et al. Anchored multiplex PCR for targeted next-generation sequencing , 2014, Nature Medicine.
[120] Yuanyuan Hou,et al. Prime editing primarily induces undesired outcomes in mice , 2020, bioRxiv.
[121] T. Lu,et al. Evaluating the Safety of Retroviral Vectors Based on Insertional Oncogene Activation and Blocked Differentiation in Cultured Thymocytes , 2016, Molecular therapy : the journal of the American Society of Gene Therapy.
[122] Matthew C. Canver,et al. Analyzing CRISPR genome-editing experiments with CRISPResso , 2016, Nature Biotechnology.
[123] 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.
[124] J. Joung,et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets , 2017, Nature Methods.