Molecular Therapy - Methods & Clinical Development

Slides:



Advertisements
Similar presentations
Molecular Therapy - Nucleic Acids
Advertisements

Targeted Disruption of V600E-Mutant BRAF Gene by CRISPR-Cpf1
Volume 24, Issue 3, Pages (March 2016)
In vivo optimization of the tagging approach using the Act5C model locus and flow cytometry-based quantification of the Act5C-GFP tagging success. In vivo.
Figure 2 Use of CRISPR/Cas9 for genome editing
Volume 12, Issue 10, Pages (September 2015)
CRISPR/Cas9-mediated genome editing of PML in human cell lines.
Volume 23, Issue 9, Pages (May 2018)
Volume 23, Issue 9, Pages (May 2018)
Transient Gene Expression by Nonintegrating Lentiviral Vectors
Molecular Therapy - Nucleic Acids
Donald B. Kohn, MD, Caroline Y. Kuo, MD 
Research Techniques Made Simple: The Application of CRISPR-Cas9 and Genome Editing in Investigative Dermatology  Joan Ramon Guitart, Jodi L. Johnson,
Mouse Genome Engineering via CRISPR-Cas9 for Study of Immune Function
Luther Davis, Nancy Maizels  Cell Reports 
Volume 7, Issue 1, Pages (April 2014)
Molecular Therapy - Nucleic Acids
Volume 13, Issue 6, Pages (December 2013)
CRISPR-Cas9 for in vivo Gene Therapy: Promise and Hurdles
Therapeutic editing of hepatocyte genome in vivo
USH2A Gene Editing Using the CRISPR System
Araksya Izmiryan, Olivier Danos, Alain Hovnanian 
Volume 7, Issue 3, Pages (September 2016)
RNA-Guided Genome Editing in Plants Using a CRISPR–Cas System
Volume 15, Issue 5, Pages (November 2014)
Andrew R. Bassett, Charlotte Tibbit, Chris P. Ponting, Ji-Long Liu 
Xiaoyu Chen, Jin Liu, Josephine M. Janssen, Manuel A.F.V. Gonçalves 
Molecular Therapy - Nucleic Acids
Volume 154, Issue 6, Pages (September 2013)
Simultaneous Reprogramming and Gene Correction of Patient Fibroblasts
Volume 1, Issue 3, Pages (September 2013)
Current Progress in Therapeutic Gene Editing for Monogenic Diseases
Volume 24, Issue 9, Pages (September 2016)
ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering
Gang Wang, Na Zhao, Ben Berkhout, Atze T Das  Molecular Therapy 
Targeted Myostatin Gene Editing in Multiple Mammalian Species Directed by a Single Pair of TALE Nucleases  Li Xu, Piming Zhao, Andrew Mariano, Renzhi.
GFP to BFP Conversion: A Versatile Assay for the Quantification of CRISPR/Cas9- mediated Genome Editing  Astrid Glaser, Bradley McColl, Jim Vadolas  Molecular.
Volume 25, Issue 11, Pages (November 2017)
Volume 2, Issue 1, Pages (January 2014)
Volume 25, Issue 11, Pages (November 2017)
Volume 25, Issue 1, Pages (January 2017)
Olivier Humbert, Nancy Maizels  Molecular Therapy - Nucleic Acids 
A CRISPR Approach to Gene Targeting
William T. Hendriks, Curtis R. Warren, Chad A. Cowan  Cell Stem Cell 
Molecular Therapy - Nucleic Acids
Molecular Therapy - Nucleic Acids
Molecular Therapy - Nucleic Acids
Increased Expression of Laminin Subunit Alpha 1 Chain by dCas9-VP160
Ignazio Maggio, Manuel A.F.V. Gonçalves  Trends in Biotechnology 
Integrase-Deficient Lentiviral Vector as an All-in-One Platform for Highly Efficient CRISPR/Cas9-Mediated Gene Editing  Pavel I. Ortinski, Bernadette.
Volume 23, Issue 3, Pages (March 2015)
Volume 19, Issue 5, Pages (May 2011)
Tagging of the endogenous Py03652 gene with the gfp gene in Plasmodium yoelii. Tagging of the endogenous Py03652 gene with the gfp gene in Plasmodium yoelii.
Molecular Therapy - Nucleic Acids
Volume 17, Issue 5, Pages (October 2016)
Ciaran M Lee, Thomas J Cradick, Gang Bao  Molecular Therapy 
CRISPR-Cas9- and AAV-mediated TCR replacement.
Gene editing by CRISPR/Cas9 for gene inactivation and targeted sequence replacement. Gene editing by CRISPR/Cas9 for gene inactivation and targeted sequence.
Volume 16, Issue 2, Pages (February 2015)
CRISPR/Cas9: Transcending the Reality of Genome Editing
Molecular Therapy - Methods & Clinical Development
Volume 9, Pages (November 2018)
Volume 13, Issue 3, Pages (October 2015)
Chemical Biology Approaches to Genome Editing: Understanding, Controlling, and Delivering Programmable Nucleases  Johnny H. Hu, Kevin M. Davis, David R.
Molecular Therapy - Methods & Clinical Development
Nuclease Target Site Selection for Maximizing On-target Activity and Minimizing Off- target Effects in Genome Editing  Ciaran M Lee, Thomas J Cradick,
Thomas Gaj, Benjamin E Epstein, David V Schaffer  Molecular Therapy 
Volume 9, Issue 4, Pages (April 2016)
Inactivation of human TRAC and CD52 genes by TALENs
Engineering Globin Gene Expression
Presentation transcript:

Molecular Therapy - Methods & Clinical Development Genome-wide Specificity of Highly Efficient TALENs and CRISPR/Cas9 for T Cell Receptor Modification  Friederike Knipping, Mark J. Osborn, Karl Petri, Jakub Tolar, Hanno Glimm, Christof von Kalle, Manfred Schmidt, Richard Gabriel  Molecular Therapy - Methods & Clinical Development  Volume 4, Pages 213-224 (March 2017) DOI: 10.1016/j.omtm.2017.01.005 Copyright © 2017 The Authors Terms and Conditions

Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions

Figure 1 DSB Repair and Targeted Genome Editing of the TCR Loci Using Designer Nucleases (A) During NHEJ-repair of TALEN- and CRISPR/Cas9-induced DSBs, frameshift mutations can result in gene knockout, or episomal IDLV can be integrated into DSBs, allowing for the permanent marking of off-target DSBs. If donor DNA is provided, HDR can lead to targeted integration of an expression cassette, i.e., therapeutic TCR chains. (B) The TCR α and β locus are composed of a number of variable (V), joining (J), constant (C), and, in the case of TRBC, diversity (D) gene segments (numbers of functional genes from IMGT/GENE-DB53 version 3.1.16, December 14, 2016). The positions of TALEN and gRNA target sequences for TCR knockout in the TCR α constant region (TRAC) and a homologous sequence shared by both TCR β constant regions (TRBC1/2) are marked by scissor symbols. DSB, DNA double-strand break; gRNA, guide RNA; HDR, homology-directed repair; IDLV, integrase-defective lentiviral vector; LTR, long terminal repeat; NHEJ, non-homologous end joining; PAM, protospacer adjacent motif; RVD, repeat variable di-residue. Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions

Figure 2 Evaluation of TALEN and CRISPR/Cas9 Activity (A) Positions of TALEN and CRISPR/Cas9 gRNA binding sites at the TRAC target site. (B) TALEN and CRISPR/Cas9 activity in the TRAC locus in K562 cells. (C) Activity of obligate heterodimeric TALENs in the TRAC locus in 293T cells. (D) TALEN and gRNA binding sites at TRBC1 and TRBC2 target locus. (E) TALEN activity in the TRBC1 and TRBC2 locus in 293T cells. (F) CRISPR/Cas9 activity in the TRBC1 and TRBC2 locus in K562 cells. (B, C, E, and F) PCR amplification of the target regions in the TCR loci produces upper bands. T7EI-mediated cleavage of NHEJ-originated heteroduplex DNA results in additional cleavage bands, marked by arrowheads. A SNP in the TRBC2 locus results in additional bands, marked by arrows (>). Ctrl, negative control; M, marker; Sp., length of spacer between TALEN binding sites in base pairs; TALENG, GoldyTALEN; TALENP(OH), pTAL3 (obligate heterodimeric FokI domain); TALENS, CAG-T7-TALEN(Sangamo)-Destination. Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions

Figure 3 Analysis of CRISPR/Cas9 and TALEN-Mediated TCR Knockout in Primary T Cells (A) Percentage of TCR knockout, evaluated by flow cytometric analysis of cell surface CD3 expression, in T cells 6 days after electroporation with αC2 or βC2. Shown are representative flow cytometry plots from four independent experiments with duplicates in T cells from three different donors. (B) Confirmation of CRISPR/Cas9 activity in T cells by T7EI assay and deep sequencing. (C) Percentage of TCR knockout, quantified by FACS, in T cells 6 days after electroporation with αT4m or βT4m. Shown are representative flow cytometry analyses of six independent experiments, with duplicates, in T cells from five different donors. (D) TALEN activity at target sites validated by T7EI assay and deep sequencing after electroporation of primary T cells with αT4m or βT4m. (B and D) Upper bands derived from wild-type PCR products and lower T7EI cleavage products are marked by arrowheads. A SNP in the TRBC1 locus results in an additional band, marked by arrows (>). The frequency of indels was analyzed by deep sequencing of PCR products. Ctrl, untreated T cells as negative control; FSC, forward scatter; M, marker; n.d., not determined. Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions

Figure 4 Quantification of NHEJ at TALEN and CRISPR/Cas9 On- and Off-Target Sites, Analyzed by Deep Sequencing and the CRISPResso Tool37 Percentage of sequences showing indels (combined insertions and deletions) at each position of the respective on- and off-target site amplicon in αT4m- and βT4m-treated primary T cells and βC1-treated K562 cells. Only indels overlapping the specified window of 20 bp that is centered on the respective predicted cleavage site are included in the quantification indicated in each graph. MM, number of mismatches to the respective target site; NC, non-coding. Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions

Figure 5 TALEN-Induced, HDR-Mediated, GFP Integration into the TRAC Locus in K562 Cells and Primary T Cells (A) Percentage of GFP-expressing K562 cells 14 days after nucleofection with GFP-encoding donor plasmid TA200G or TA800G without (upper panel) or together with TALEN αT4P or βT4P (lower panel). Representative FACS analyses of two independent experiments are shown. (B) Validation of targeted integration into the TRAC locus by bidirectional PCR with primers binding in the GFP expression cassette or outside of the 5′ or 3′ homology arms, respectively. (C) Flow cytometric analysis of TCR knockout, determined by CD3 surface expression, and GFP expression of primary T cells 7, 15, and 21 days after transduction with TA800G-IDLV only (right panels) or additional electroporation with αT4m on day 1 (left panels). Shown are representative FACS analyses of three independent experiments in T cells from separate donors. (D) Validation of targeted integration into the TRAC locus by bidirectional PCR with primers binding in the GFP expression cassette or outside of the 5′ or 3′ homology arm, respectively (see scheme in B). PCR products are marked by arrowheads. Hom., homology region. Molecular Therapy - Methods & Clinical Development 2017 4, 213-224DOI: (10.1016/j.omtm.2017.01.005) Copyright © 2017 The Authors Terms and Conditions