gene editing: paving the way for accelerated clinical ......crispr/cas9 and zinc finger nuclease...

1
Precision genome engineering requires technologies that allow efficient and reproducible delivery of DNA, mRNA and RNP-based reagents into a range of primary cells and stem cells. In addition, clinical gene editing requires a transfection platform that is GMP-compliant and scalable to accommodate billions of cells in a single transfection. Here we share data on gene deletion and correction in primary cells and cell lines following transfection of CRISPR/cas9 and zinc finger nuclease molecules using MaxCyte’s clinically validated flow electroporation platform. Data on efficient modification of induced pluripotent stem cells, knockdown of checkpoint inhibitors in T cells and correction of point mutations in hematopoietic stem cells are included along with examples of process scalability. Finally, we include data on multiplex editing and gene knockout in T- and NK-cells. Abstract Gene Editing: Paving the Way for Accelerated Clinical Development of Adoptive Cell Immunotherapies. Peer Heine, PhD, Peter Gee, PhD, Pachai Natarajan, PhD and James Brady PhD. MaxCyte, Gaithersburg, MD, USA Corresponding Author: James Brady; [email protected] MaxCyte, Inc., Tel: (301) 944-1700 [email protected] , www.maxcyte.com Flow Electroporation, MaxCyte, ExPERT and MaxCyte GT are trademarks of MaxCyte, Inc.. Case Study #1 Dual Knockout of TRAC & β2-microglobulin in Activated T Cells Optimization of Cas9 RNP and sgRNA Delivery Produces >80% Double Knockout Figure 1: Optimization of RNP, sgRNA Delivery for CRISPR-mediated Dual Knockdown of T Cell Receptor (CD3) and HLA. A) Primary activated T cells were electroporation with Cas9 ribonucleoprotein (RNP) and sgRNAs targeting TRAC and β2-microglobulin using the MaxCyte GT using two different electroporation protocols and three different concentrations of RNP and sgRNAs. B) Cell viability and the rate of cell expansion were assessed for 18 days following electroporation. C) The percent dual knockout efficiency was determined using FACS 12 days post electroporation. Cells electroporated using protocol 1 with 10 µg of Cas9 RNP and 2 µg each of TRAC- and β2-microglobulin sgRNAs displayed an 84% dual knockout. C. HLA/CD3 Knockout Analysis B. Cell Viability & Recovery Time A. Optimization Parameters Summary Gene editing using MaxCyte non-viral engineering enables rapid development of next-generation adoptive cell therapies for treatment of a wide variety of diseases The high viability of cells following electroporation allows for high rates of long-term engraftment that are required for patient treatment Flow Electroporation ® Technology efficiently (co)delivers a diversity of payloads including mRNA, sgRNA, and RNPs to difficult-to-engineer primary cells commonly used for adoptive cell therapies including hematopoietic stem cells and T cells The high efficiency and low toxicity of Flow Electroporation provide for high levels of nuclease or nickase-mediated gene editing including: - Gene knockout/disruption - Gene knock-in - Single nucleotide gene mutation correction MaxCyte clinical scalability and regulatory compliance provide for streamlined clinical translation of new therapies Case Study #2 Checkpoint Inhibitor Knockout Using Paired Cas9 Nickases High Efficiency Nickase-mediated Gene Knockout in Human Primary Hematopoietic Cells Figure 2: Paired Nickase RNPs Outperform spCas9 RNPs for Checkpoint Inhibitor Knockout in Human Primary Hematopoietic Cells. In a paired nickase approach, Streptococcus pyogenes Cas9 (SpCas9) is modified to inactivate one of its two nuclease domains, causing single-stranded nicks rather than a double stranded break. Paired with a second Cas9 nickase targeting a proximal location, these two single-stranded nicks cause a DSB to occur. Off-target modifications caused by paired gRNA are rare. A) Human primary T cells were electroporated with paired SpCas9 nickase RNP, with or without PD-1-specific sgRNAs. Genomic DNAs were harvested 72 hours post-electroporation for indels analysis. B) Human primary T cells were electroporated with SpCas9 RNP or paired nickase RNP with or without PD-1-specific sgRNA. At various times post electroporation, cells were assessed via FACS for surface PD-1 expression. C) Human primary CD8+ T cells were electroporated withSpCas9 RNP or paired SpCas9 nickase RNP specific for CTLA4 or TIM-3. Genomic DNA was harvested 120 hours post-electroporation for indels analysis. A C: Averages from three biological replicates are plotted with error bars representing one standard deviation. bioRxiv Sept. 2019 preprint (https://doi.org/10.1101/766493). A. Nickase-mediated Knockout of PD-1 in Primary T Cells B. Gene Knockout Correlates With Lower PD-1 Expression C. Multiple Target Knockout in Primary CD8 + T Cells Case Study #3 Ex Vivo Gene Correction in X-CGD Patient Stem Cells Clinically-relevant Levels of gp91+ Cells in the Peripheral Blood and Bone Marrow Following Engraftment Background: X-linked chronic granulomatous disease (X-CGD) is caused by a single nucleotide mutation in the CYBB gene which encodes a critical component (gp91-phox) of NADPH oxidase, an enzyme that is key for the anti-microbial activity of phagocytes. Correction of mutation within the faulty CYBB gene offers a new curative treatment for X-CGD patients. The patients’ own cells are harvested, the mutated gene corrected using CRISPR-mediated gene editing, and the cells with the corrected gene returned to the patient. The engrafted cells multiply to create a new population of cells displaying ‘normal’ function and eliminating disease. Figure 3: Efficient gp91+ Mutation Correction with Restored NOX2 Activity 20 Weeks Post Engraftment. CD34+ hematopoietic stem cells (HSPCs) were isolated from X-CGD patients and electroporated with CRISPR/Cas9, guide RNA, and the gene correcting oligo template using the MaxCyte GT. A) A portion of the cells were differentiated in vitro into myeloid cells and gene correction rates determined to be 31%. The other portion of corrected HSPCs were introduced into immunodeficient mice. After 20 weeks the engrafted human cells in the mouse peripheral blood (B) expressed the corrected gp91 gene at 21%, while the engrafted cells in the bone marrow (C) showed a 34% gp91 expression with 10% displaying NOX2 activity as assessed by the DHR assay (D). These correction rates are within clinically beneficial potency thresholds. Sci. Transl. Med., 9(372), Jan 2017. X-CGD Patient CRISPR/Cas9 + gRNA + Oligo Template hCD34+ HSPCs NOD-SCIDc-/- Mouse Engraftment In vitro Myeloid Differentiation Gp91 Gene Correction Electroporated Patient Cells Untreated Patient Cells Untreated Healthy Cells 31% A. Restored gp91 Expression in Differentiated Myeloid Cells B. gp91 Expression in Mouse Peripheral Blood 20 weeks post Engraftment Healthy Cells Electroporated Patient Cells Healthy Cells Electroporated Patient Cells C. gp91 Expression in Mouse Bone Marrow 20 weeks post Engraftment D. NOX2 Activity in Mouse Bone Marrow 20 weeks post Engraftment Figure 4: High Biallelic CCR5 Gene Disruption Rates Using ZFN mRNA Delivery With Ability for Long-term Engraftment. Researchers at Sangamo developed a CCR5-targeted zinc finger nuclease that they showed was active in a variety of CD4 T cells and HSPCs and that conferred resistance to HIV infection. This therapy was advanced to the clinic using adenoviruses to deliver the ZFN constructs. The phase I/II trials showed that CD4 cells with a disrupted CCR5 gene could be engrafted, were safe and persisted. Toxicity related to the adenoviral vector precluded the intended trials from progressing. To rescue the therapy, the company turned to mRNA delivery of the CCR5-specific ZFN using the MaxCyte GT. The work published in Mol Ther. Methods Clin. Dev., 3, 2016 and summarized above demonstrate the rapid progression from process development of ZFN delivery, through manufacturing qualification runs, pre-clinical toxicity studies and initiation of clinical trial NCT02500849 using the MaxCyte GT. See publication for detailed methods. Case Study #4 Advancing an HIV Clinical Program for CCR5 Gene Disruption Rapid Development & IND-enabling Pre-clinical Studies Support Progression of Clinical Trial Cryopreserved Cells from GLP Runs from 3 Independent Human Donors 20 weeks 18-31% CCR5 disruption in hCD45+ cells in bone marrow 10-20% CCR5 disruption in hCD45+ PBMCs NSG Mouse Engraftment No difference in premature deaths No adverse events No toxicity associated with modified cells >80% Viability 54-67% CCR5 Disruption 20 weeks 20 weeks Observations CCR5 Gene Disruption Rate vs. [mRNA] Bulk Pools Cell Viability vs. [mRNA] Bulk Pools Day 1 post EP Day 2 post EP No impact on ability for long- term engraftment Healthy Human Donor CCR5-targeted ZFN mRNA (rSB-728mR) hCD34+ HSPCs Bulk Pool Analysis Individual Colony Forming Unit (CFU) Analysis CCR5 Gene Disruption in Individual CFU No impact on hematopoietic potential Full-scale Engineering Runs Using Different Human Donors & Toxicity Testing Customer Data

Upload: others

Post on 24-Aug-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Gene Editing: Paving the Way for Accelerated Clinical ......CRISPR/cas9 and zinc finger nuclease molecules using MaxCyte’sclinically validated flow electroporation platform. Data

Precision genome engineering requires technologies that allow efficient and reproducible delivery of DNA, mRNA and RNP-based reagents into a range of primary cells and stem cells. In addition, clinical gene editing requires atransfection platform that is GMP-compliant and scalable to accommodate billions of cells in a single transfection. Here we share data on gene deletion and correction in primary cells and cell lines following transfection ofCRISPR/cas9 and zinc finger nuclease molecules using MaxCyte’s clinically validated flow electroporation platform. Data on efficient modification of induced pluripotent stem cells, knockdown of checkpoint inhibitors in T cells andcorrection of point mutations in hematopoietic stem cells are included along with examples of process scalability. Finally, we include data on multiplex editing and gene knockout in T- and NK-cells.

Abstract

Gene Editing: Paving the Way for Accelerated Clinical Development of Adoptive Cell Immunotherapies.Peer Heine, PhD, Peter Gee, PhD, Pachai Natarajan, PhD and James Brady PhD. MaxCyte, Gaithersburg, MD, USA

Corresponding Author: James Brady; [email protected] MaxCyte, Inc., Tel: (301) 944-1700 [email protected], www.maxcyte.com

Flow Electroporation, MaxCyte, ExPERT and MaxCyte GT are trademarks of MaxCyte, Inc..

Case Study #1 – Dual Knockout of TRAC & β2-microglobulin in Activated T CellsOptimization of Cas9 RNP and sgRNA Delivery Produces >80% Double Knockout

Figure 1: Optimization of RNP, sgRNA Delivery for CRISPR-mediated Dual Knockdown of T Cell Receptor (CD3) and HLA. A)Primary activated T cells were electroporation with Cas9 ribonucleoprotein (RNP) and sgRNAs targeting TRAC and β2-microglobulinusing the MaxCyte GT using two different electroporation protocols and three different concentrations of RNP and sgRNAs. B) Cellviability and the rate of cell expansion were assessed for 18 days following electroporation. C) The percent dual knockout efficiencywas determined using FACS 12 days post electroporation. Cells electroporated using protocol 1 with 10 µg of Cas9 RNP and 2 µgeach of TRAC- and β2-microglobulin sgRNAs displayed an 84% dual knockout.

C. HLA/CD3 Knockout Analysis

B. Cell Viability & Recovery Time

A. Optimization Parameters

Summary• Gene editing using MaxCyte non-viral engineering enables rapid development of next-generation

adoptive cell therapies for treatment of a wide variety of diseases

• The high viability of cells following electroporation allows for high rates of long-term engraftment that are required for patient treatment

• Flow Electroporation® Technology efficiently (co)delivers a diversity of payloads including mRNA, sgRNA, and RNPs to difficult-to-engineer primary cells commonly used for adoptive cell therapies including hematopoietic stem cells and T cells

• The high efficiency and low toxicity of Flow Electroporation provide for high levels of nuclease or nickase-mediated gene editing including:

- Gene knockout/disruption

- Gene knock-in

- Single nucleotide gene mutation correction

• MaxCyte clinical scalability and regulatory compliance provide for streamlined clinical translation of new therapies

Case Study #2 – Checkpoint Inhibitor Knockout Using Paired Cas9 NickasesHigh Efficiency Nickase-mediated Gene Knockout in Human Primary Hematopoietic Cells

Figure 2: Paired Nickase RNPs Outperform spCas9 RNPs for Checkpoint Inhibitor Knockout in Human Primary HematopoieticCells. In a paired nickase approach, Streptococcus pyogenes Cas9 (SpCas9) is modified to inactivate one of its two nucleasedomains, causing single-stranded nicks rather than a double stranded break. Paired with a second Cas9 nickase targeting a proximallocation, these two single-stranded nicks cause a DSB to occur. Off-target modifications caused by paired gRNA are rare. A) Humanprimary T cells were electroporated with paired SpCas9 nickase RNP, with or without PD-1-specific sgRNAs. Genomic DNAs wereharvested 72 hours post-electroporation for indels analysis. B) Human primary T cells were electroporated with SpCas9 RNP orpaired nickase RNP with or without PD-1-specific sgRNA. At various times post electroporation, cells were assessed via FACS forsurface PD-1 expression. C) Human primary CD8+ T cells were electroporated withSpCas9 RNP or paired SpCas9 nickase RNPspecific for CTLA4 or TIM-3. Genomic DNA was harvested 120 hours post-electroporation for indels analysis. A – C: Averages fromthree biological replicates are plotted with error bars representing one standard deviation. bioRxiv Sept. 2019preprint (https://doi.org/10.1101/766493).

A. Nickase-mediated Knockout of PD-1 in Primary T Cells

B. Gene Knockout Correlates With Lower PD-1 Expression

C. Multiple Target Knockout in Primary CD8+ T Cells

Case Study #3 – Ex Vivo Gene Correction in X-CGD Patient Stem CellsClinically-relevant Levels of gp91+ Cells in the Peripheral Blood and Bone Marrow Following Engraftment

Background: X-linked chronic granulomatous disease (X-CGD) is caused by a single nucleotide mutation in the CYBB gene which encodes a critical component (gp91-phox) of NADPH oxidase, an enzyme that is key for the anti-microbial activity of phagocytes. Correction of mutation within the faulty CYBB gene offers a new curative treatment for X-CGD patients. The patients’ own cells are harvested, the mutated gene corrected using CRISPR-mediated gene editing, and the cells with the corrected gene returned to the patient. The engrafted cells multiply to create a new population of cells displaying ‘normal’function and eliminating disease.

Figure 3: Efficient gp91+ Mutation Correction with Restored NOX2 Activity 20 Weeks Post Engraftment. CD34+ hematopoieticstem cells (HSPCs) were isolated from X-CGD patients and electroporated with CRISPR/Cas9, guide RNA, and the gene correctingoligo template using the MaxCyte GT. A) A portion of the cells were differentiated in vitro into myeloid cells and gene correctionrates determined to be 31%. The other portion of corrected HSPCs were introduced into immunodeficient mice. After 20 weeks theengrafted human cells in the mouse peripheral blood (B) expressed the corrected gp91 gene at 21%, while the engrafted cells in thebone marrow (C) showed a 34% gp91 expression with 10% displaying NOX2 activity as assessed by the DHR assay (D). Thesecorrection rates are within clinically beneficial potency thresholds. Sci. Transl. Med., 9(372), Jan 2017.

X-CGD Patient

CRISPR/Cas9 + gRNA + Oligo

Template

hCD34+ HSPCs

NOD-SCIDꙋc-/-

Mouse Engraftment

In vitro Myeloid Differentiation

Gp91 Gene

Correction

ElectroporatedPatient Cells

Untreated Patient Cells

Untreated Healthy Cells

31%

A. Restored gp91 Expression in Differentiated Myeloid Cells

B. gp91 Expression in Mouse Peripheral Blood – 20 weeks post Engraftment

Healthy CellsElectroporatedPatient Cells

Healthy CellsElectroporatedPatient Cells

C. gp91 Expression in Mouse Bone Marrow – 20 weeks post Engraftment

D. NOX2 Activity in Mouse Bone Marrow – 20 weeks post Engraftment

Figure 4: High Biallelic CCR5 Gene Disruption Rates Using ZFN mRNA Delivery With Ability for Long-term Engraftment.Researchers at Sangamo developed a CCR5-targeted zinc finger nuclease that they showed was active in a variety of CD4 T cells andHSPCs and that conferred resistance to HIV infection. This therapy was advanced to the clinic using adenoviruses to deliver the ZFNconstructs. The phase I/II trials showed that CD4 cells with a disrupted CCR5 gene could be engrafted, were safe and persisted.Toxicity related to the adenoviral vector precluded the intended trials from progressing. To rescue the therapy, the company turnedto mRNA delivery of the CCR5-specific ZFN using the MaxCyte GT. The work published in Mol Ther. Methods Clin. Dev., 3, 2016 andsummarized above demonstrate the rapid progression from process development of ZFN delivery, through manufacturingqualification runs, pre-clinical toxicity studies and initiation of clinical trial NCT02500849 using the MaxCyte GT. See publication fordetailed methods.

Case Study #4 – Advancing an HIV Clinical Program for CCR5 Gene DisruptionRapid Development & IND-enabling Pre-clinical Studies Support Progression of Clinical Trial

Cryopreserved Cells from GLP Runs from 3 Independent Human Donors

20 weeks

18-31%CCR5 disruption in hCD45+

cells in bone marrow

10-20%CCR5 disruption in

hCD45+ PBMCs

NSG Mouse Engraftment

• No difference in premature deaths

• No adverse events

• No toxicity associated with modified cells

>80%Viability

54-67%CCR5 Disruption

20 weeks

20 weeks

Observations

CCR5 Gene Disruption Rate vs. [mRNA] Bulk Pools

Cell Viability vs. [mRNA] Bulk Pools

Day 1 post EP Day 2 post EP

• No impact on ability for long-term engraftment

Healthy Human Donor

CCR5-targeted ZFN mRNA (rSB-728mR)

hCD34+ HSPCs

Bulk Pool Analysis

Individual Colony Forming Unit (CFU) Analysis

CCR5 Gene Disruption in Individual CFU

• No impact on hematopoietic potential

Full-scale Engineering Runs Using Different Human Donors & Toxicity Testing

Customer Data