scholarly journals The clinical applications of genome editing in HIV

Blood ◽  
2016 ◽  
Vol 127 (21) ◽  
pp. 2546-2552 ◽  
Author(s):  
Cathy X. Wang ◽  
Paula M. Cannon

Abstract HIV/AIDS has long been at the forefront of the development of gene- and cell-based therapies. Although conventional gene therapy approaches typically involve the addition of anti-HIV genes to cells using semirandomly integrating viral vectors, newer genome editing technologies based on engineered nucleases are now allowing more precise genetic manipulations. The possible outcomes of genome editing include gene disruption, which has been most notably applied to the CCR5 coreceptor gene, or the introduction of small mutations or larger whole gene cassette insertions at a targeted locus. Disruption of CCR5 using zinc finger nucleases was the first-in-human application of genome editing and remains the most clinically advanced platform, with 7 completed or ongoing clinical trials in T cells and hematopoietic stem/progenitor cells (HSPCs). Here we review the laboratory and clinical findings of CCR5 editing in T cells and HSPCs for HIV therapy and summarize other promising genome editing approaches for future clinical development. In particular, recent advances in the delivery of genome editing reagents and the demonstration of highly efficient homology-directed editing in both T cells and HSPCs are expected to spur the development of even more sophisticated applications of this technology for HIV therapy.

2017 ◽  
Vol 37 (01) ◽  
pp. 45-52 ◽  
Author(s):  
Simone Haas ◽  
Viviane Dettmer ◽  
Toni Cathomen

SummaryTargeted genome editing with designer nucleases, such as zinc finger nucleases, TALE nucleases, and CRISPR-Cas nucleases, has heralded a new era in gene therapy. Genetic disorders, which have not been amenable to conventional gene-addition-type gene therapy approaches, such as disorders with dominant inheritance or diseases caused by mutations in tightly regulated genes, can now be treated by precise genome surgery. Moreover, engineered nucleases enable novel genetic interventions to fight infectious diseases or to improve cancer immunotherapies. Here, we review the development of the different classes of programmable nucleases, discuss the challenges and improvements in translating gene editing into clinical use, and give an outlook on what applications can expect to enter the clinic in the near future.


2021 ◽  
Vol 11 ◽  
Author(s):  
Satya Swathi Nadakuduti ◽  
Felix Enciso-Rodríguez

The year 2020 marks a decade since the first gene-edited plants were generated using homing endonucleases and zinc finger nucleases. The advent of CRISPR/Cas9 for gene-editing in 2012 was a major science breakthrough that revolutionized both basic and applied research in various organisms including plants and consequently honored with “The Nobel Prize in Chemistry, 2020.” CRISPR technology is a rapidly evolving field and multiple CRISPR-Cas derived reagents collectively offer a wide range of applications for gene-editing and beyond. While most of these technological advances are successfully adopted in plants to advance functional genomics research and development of innovative crops, others await optimization. One of the biggest bottlenecks in plant gene-editing has been the delivery of gene-editing reagents, since genetic transformation methods are only established in a limited number of species. Recently, alternative methods of delivering CRISPR reagents to plants are being explored. This review mainly focuses on the most recent advances in plant gene-editing including (1) the current Cas effectors and Cas variants with a wide target range, reduced size and increased specificity along with tissue specific genome editing tool kit (2) cytosine, adenine, and glycosylase base editors that can precisely install all possible transition and transversion mutations in target sites (3) prime editing that can directly copy the desired edit into target DNA by search and replace method and (4) CRISPR delivery mechanisms for plant gene-editing that bypass tissue culture and regeneration procedures including de novo meristem induction, delivery using viral vectors and prospects of nanotechnology-based approaches.


Blood ◽  
2016 ◽  
Vol 127 (21) ◽  
pp. 2536-2545 ◽  
Author(s):  
Matthew C. Canver ◽  
Stuart H. Orkin

Abstract Despite nearly complete understanding of the genetics of the β-hemoglobinopathies for several decades, definitive treatment options have lagged behind. Recent developments in technologies for facile manipulation of the genome (zinc finger nucleases, transcription activator-like effector nucleases, or clustered regularly interspaced short palindromic repeats–based nucleases) raise prospects for their clinical application. The use of genome-editing technologies in autologous CD34+ hematopoietic stem and progenitor cells represents a promising therapeutic avenue for the β-globin disorders. Genetic correction strategies relying on the homology-directed repair pathway may repair genetic defects, whereas genetic disruption strategies relying on the nonhomologous end joining pathway may induce compensatory fetal hemoglobin expression. Harnessing the power of genome editing may usher in a second-generation form of gene therapy for the β-globin disorders.


2019 ◽  
Author(s):  
Geoffrey L. Rogers ◽  
Hsu-Yu Chen ◽  
Heidy Morales ◽  
Paula M. Cannon

AbstractAdeno-associated virus (AAV) vectors are frequently used as donor templates for genome editing by homologous recombination. Although modification rates are typically under 1%, they are greatly enhanced by targeted double-stranded DNA breaks (DSBs). A recent report described clade F AAVs mediating high-efficiency homologous recombination-based editing in the absence of DSBs. The clade F vectors included AAV9 and a series isolated from human hematopoietic stem/progenitor cells (HSPCs). We evaluated these vectors by packaging homology donors into AAV9 and an AAVHSC capsid and examining their ability to insert GFP at the CCR5 or AAVS1 loci in human HSPCs and cell lines. As a control we used AAV6, which effectively edits HSPCs, but only when combined with a targeted DSB. Each AAV vector promoted GFP insertion in the presence of matched CCR5 or AAVS1 zinc finger nucleases (ZFNs), but none supported detectable editing in the absence of the nucleases. Rates of editing with ZFNs correlated with transduction efficiencies for each vector, implying no differences in the ability of donor sequences delivered by the different vectors to direct genome editing. Our results therefore do not support that clade F AAVs can perform high efficiency genome editing in the absence of a DSB.


2021 ◽  
Vol 28 (2) ◽  
pp. 8
Author(s):  
Gunda Petraitytė ◽  
Eglė Preikšaitienė ◽  
Violeta Mikštienė

Studies which seek fundamental, thorough knowledge of biological processes, and continuous advancement in natural sciences and biotechnology enable the establishment of molecular strategies and tools to treat disorders caused by genetic mutations. Over the years biological therapy evolved from using stem cells and viral vectors to RNA therapy and testing different genome editing tools as promising gene therapy agents. These genome editing technologies (Zinc finger nucleases, TAL effector nucleases), specifically CRISPR-Cas system, revolutionized the field of genetic engineering and is widely applied to create cell and animal models for various hereditary, infectious human diseases and cancer, to analyze and understand the molecular and cellular base of pathogenesis, to find potential drug/treatment targets, to eliminate pathogenic DNA changes in various medical conditions and to create future “precise medication”. Although different concerning factors, such as precise system delivery to the target cells, efficacy and accuracy of editing process, different approaches of making the DNA changes as well as worrying bioethical issues remain, the importance of genome editing technologies in medicine is undeniable. The future of innovative genome editing approach and strategies to treat diseases is complicated but interesting and exciting at once for all related parties – researchers, clinicians, and patients.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi127-vi127
Author(s):  
Bindu Hegde ◽  
Theodore Roth ◽  
David Nguyen ◽  
Zinal Chheda ◽  
Ryan Apathy ◽  
...  

Abstract We recently identified an HLA-A*02:01-restricted CD8 T cell epitope encompassing the H3.3K27M mutation, which is common in diffuse midline glioma, and a corresponding high-affinity T cell receptor (TCR) that recognizes the epitope. While recombinant viral vectors have been widely used for genetic reprogramming of T cells, viral vectors are far from ideal as they typically integrate randomly into the genome and are not governed by the molecular regulatory mechanisms of the cell. We used a non-viral, CRISPR-Cas9-based approach to replace the endogenous TCR with H3.3K27M TCR at the TCR a constant region (TRAC) in human T cells. Co-electroporation of healthy donor-derived T cells with homology-directed repair (HDR) templates encoding the full-length sequence of H3.3K27M TCR along with CRISPR-Cas9 ribonucleoprotein (RNP) resulted in the integration of the new TCR into the TRAC locus by HDR. Antibody staining of TCR α/β and H3.3K27M dextramer showed replacement of endogenous TCR with H.3.3K27M TCR in ~5–10% of TCR+ CD8 T cells. Modifying the HDR template to include a binding site for Cas9, which contains the nuclear localization signal that acts as a “shuttle”, further enhanced the integration efficiency (~15% of TCR+ CD8 T cells). Furthermore, HLA-A2+ H3.3K27M TCR-engineered T cells selectively killed U87 glioma cells expressing the H3.3K27M epitope. In addition, the engineered T cells exhibited a stem memory-like phenotype when expanded in the presence of a cocktail of IL-2, IL-7 and IL-15. Taken together, these data provide evidence for non-viral genome editing as a strategy to engineer T cells with specific TCR for cancer immunotherapy.


Sign in / Sign up

Export Citation Format

Share Document