Fact-checked by Grok 2 weeks ago

Genome editing

Genome editing encompasses methods for precisely altering DNA sequences in living organisms, enabling targeted insertions, deletions, replacements, or modifications of genetic material to study or treat diseases. These techniques rely on engineered nucleases that create double-strand breaks at specific genomic loci, which are then repaired by cellular mechanisms such as or , often incorporating desired changes. Early approaches included zinc-finger nucleases (ZFNs) developed in the and transcription activator-like effector nucleases (TALENs) in the , but the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) system, adapted from bacterial adaptive immunity and demonstrated for eukaryotic genome editing in 2012, has dominated due to its simplicity, efficiency, and versatility. Significant achievements include rapid gene knockout in model organisms for functional genomics, crop improvement for enhanced yield and resistance, and therapeutic applications in humans, such as the 2023 FDA approval of CRISPR-based ex vivo editing for sickle cell disease and beta-thalassemia, marking the first clinical use of the technology to correct pathogenic mutations. These advances stem from empirical validation of editing precision in controlled trials, demonstrating causal links between corrected genotypes and phenotypic improvements without widespread off-target effects in targeted contexts. However, challenges persist, including unintended mutations from incomplete specificity and delivery inefficiencies in vivo, necessitating ongoing refinements like base editing and prime editing for higher fidelity. Controversies arise primarily from editing, where changes are heritable, raising concerns over safety risks like mosaicism and long-term ecological impacts, as well as ethical debates on and inequality, though applications avoid inheritance issues and focus on individual therapeutic benefits supported by clinical data. The 2018 case of unauthorized human embryo editing by highlighted regulatory gaps but also underscored the technology's potential when responsibly applied, with favoring paused heritable use until risks are empirically mitigated.

History

Pre-Engineered Nuclease Era

Homologous recombination (HR), a conserved cellular mechanism for repairing double-strand breaks using a homologous DNA template, formed the basis of early genome editing efforts before the advent of engineered site-specific nucleases. In this era, spanning the 1970s to late 1980s, researchers introduced linear DNA constructs with flanking homology arms matching the target genomic locus, relying on spontaneous, low-frequency HR events to achieve precise insertions, deletions, or replacements without inducing targeted DNA breaks. This approach yielded high-fidelity modifications when successful but suffered from extreme inefficiency, typically 10^{-4} to 10^{-6} in mammalian cells, dominated by random non-homologous integrations. To counter this, positive-negative selection strategies were developed, using markers like neomycin resistance for enrichment and herpes simplex virus thymidine kinase for counterselection against random integrants. Pioneering work in , where is naturally more efficient, demonstrated feasibility early on. In 1979, Scherer and Davis achieved targeted chromosomal segment replacement in by transfecting hybrid plasmids, marking one of the first instances of precise genomic alteration via . This success in unicellular eukaryotes informed extensions to mammals. advanced the field in 1985 by reporting -mediated insertion of a functional gene into the human beta-globin locus in cultured erythroleukemia cells, confirming targeted events at frequencies around 1 in 10^3 to 10^4 transformants under selection. The integration of mouse embryonic stem (ES) cells, first isolated in 1981 by Martin Evans and Matthew Kaufman, enabled heritable modifications. By 1987, Smithies and independently applied in mouse ES cells to specific genes, such as Aph-3, using isogenic targeting vectors to boost efficiency. Capecchi's group refined selection protocols, achieving targeted disruptions at rates improved to about 1 in 10^5 electroporated cells. These methods culminated in the first germline-transmissible gene s in mice by 1989, allowing systematic functional analysis of genes via "" models. Despite these advances, the absence of DSB induction limited scalability, confining applications largely to tractable systems like and mouse ES cells for into gene function and disease modeling.

Development of Site-Specific Nucleases

Site-specific nucleases emerged as tools for targeted genome editing by inducing double-strand breaks (DSBs) at predetermined DNA sequences, leveraging cellular pathways for precise modifications. Early efforts focused on meganucleases, naturally occurring homing endonucleases from microbes with recognition sites of 12-40 base pairs, first characterized in the 1980s such as I-SceI from mitochondria. These enzymes demonstrated enhanced gene targeting via DSB-stimulated in and mammalian cells by the early 1990s, but their rigid protein-DNA interfaces limited redesign for new specificities, restricting broad applicability. Engineering attempts in the 2000s involved semi-rational design and to alter specificity, yet success remained low due to coupled recognition and cleavage domains. To overcome meganuclease limitations, researchers developed modular nucleases by fusing customizable DNA-binding domains to separate, non-specific nuclease modules. nucleases (ZFNs), invented in 1996 by fusing proteins—discovered in 1985—with the endonuclease cleavage domain, enabled programmable targeting of 9-18 sites as dimers. Initial demonstrations achieved targeted DSBs and gene disruption in mammalian cells by 1998, with therapeutic applications emerging in the 2000s, including HIV resistance via CCR5 knockout in human cells. However, ZFNs required expertise in zinc finger assembly, often via or structure-based design, and off-target effects arose from context-dependent binding affinities. Transcription activator-like effector nucleases (TALENs) advanced programmability in 2010, following the 2009 deciphering of the TALE DNA-binding code from bacteria, where repeat-variable di-residues (RVDs) specify one each. TALENs pair FokI domains flanking a central spacer for dimerization and cleavage, offering higher specificity than ZFNs due to independent modular recognition and reduced toxicity in applications like and human cell . First used for targeted knockouts and insertions in 2011, TALENs facilitated multiplex and expanded genome engineering to non-model organisms, though assembly of lengthy TALE arrays remained labor-intensive compared to later RNA-guided methods. These innovations established DSB-based principles, paving the way for scalable technologies while highlighting trade-offs in specificity, ease of design, and delivery.

CRISPR Breakthrough and Rapid Adoption

The CRISPR-Cas9 system emerged as a transformative tool for genome editing following a 2012 study by and , who demonstrated that the Cas9 endonuclease from , guided by a dual structure (crRNA and tracrRNA, later simplified into single-guide RNA), could be reprogrammed to induce site-specific double-strand breaks in DNA. This work, published online on June 28, 2012, in Science, repurposed the bacterial adaptive immune mechanism—previously characterized in the early 2000s—for precise targeting, offering advantages in simplicity, multiplexing potential, and cost over prior nucleases like ZFNs and TALENs. The system's -guided specificity stemmed from base-pairing between the guide RNA and target DNA, adjacent to a protospacer-adjacent motif (PAM) sequence, enabling predictable cleavage without for each target. Adaptation to cellular genome editing occurred rapidly, with demonstrations in prokaryotic and eukaryotic systems by early 2013. Independent studies by Feng Zhang's group at the Broad Institute and George Church's lab at Harvard achieved targeted modifications via (NHEJ) and (HDR) in and lines, as reported in Science on January 3, 2013. These applications exploited the endogenous pathways to introduce insertions, deletions, or precise substitutions, validating CRISPR-Cas9's efficiency in mammalian genomes where off-target effects, though present, were manageable compared to earlier tools. Concurrently, Virginijus Šikšnys's group in reported similar prokaryotic editing, underscoring the technology's versatility. Adoption accelerated exponentially, evidenced by a surge in research output: CRISPR-related publications rose from fewer than 100 annually pre-2012 to over 3,900 by 2018, reflecting its integration into diverse fields like functional genomics and model organism engineering. Patent filings intensified, with the University of California (representing Doudna's work) submitting the first provisional application in May 2012, followed by the Broad Institute's December 2012 filing—expedited to yield the initial U.S. patent in April 2014 for eukaryotic applications—sparking ongoing interference proceedings that highlighted competing claims but did not impede lab proliferation. By 2015, CRISPR had supplanted prior methods in most academic and industrial workflows due to its accessibility, enabling high-throughput screens and multiplexed edits unattainable with protein-based nucleases.

Post-2020 Advancements and Commercialization

Following the rapid adoption of CRISPR- in the late , post-2020 developments emphasized enhanced precision, reduced off-target effects, and delivery to expand therapeutic applicability. Researchers introduced refined variants, such as smaller Cas12 and Cas13 orthologs, enabling better packaging into viral vectors for and multiplex capabilities. These variants improved editing efficiency in non-dividing cells, addressing limitations in earlier systems. Base editing and prime editing matured as DSB-free alternatives, minimizing unwanted insertions or deletions. Base editors, which convert specific via deaminase fusion, entered clinical trials post-2020 for conditions like , with early 2025 data showing successful single-base corrections in human subjects. , leveraging a reverse transcriptase-pegsRNA complex, advanced to support diverse modifications including insertions up to dozens of bases, with preclinical models demonstrating up to 50% efficiency in therapeutically relevant genes by 2025. These tools expanded the editable genome fraction beyond traditional CRISPR's reach. Delivery innovations accelerated in vivo applications, with nanoparticle and lipid-conjugated systems achieving tissue-specific targeting, as shown in 2024 studies editing subsets of neurons or hepatocytes in animal models without broad toxicity. Clinical translation progressed, with over 15 base-editing trials registered by mid-2025 targeting immunodeficiencies and metabolic disorders. Commercialization gained momentum with regulatory approvals validating ex vivo CRISPR therapies. In December 2023, the FDA approved Casgevy (exagamglogene autotemcel), a CRISPR-Cas9-edited autologous from and , for in patients aged 12 and older with recurrent vaso-occlusive crises; approval for transfusion-dependent followed in January 2024 for those aged 12 and older. Casgevy disrupts the BCL11A enhancer to reactivate , achieving transfusion independence in 94% of beta thalassemia patients and reducing vaso-occlusive events by 91% in sickle cell cases across trials. This marked the first CRISPR-based therapy commercialization, though high costs exceeding $2 million per treatment and manufacturing complexities limited initial access. By 2025, the sector saw expanded pipelines, with prioritizing programs for cardiovascular and autoimmune diseases, alongside Phase 3 trials for . The global CRISPR gene- market reached $4.01 billion in 2024, projected to grow to $13.50 billion by 2033, driven by diagnostics, , and therapeutics, though disputes and ethical concerns over editing persisted. Ongoing trials numbered over 50 worldwide, focusing on , , and rare diseases, signaling broader clinical maturation.

Biological and Mechanistic Foundations

DNA Repair Mechanisms Exploited in Editing

Genome editing technologies, such as those employing site-specific nucleases, induce double-strand breaks (DSBs) in DNA that are subsequently repaired by cellular pathways, enabling targeted genetic modifications. The primary pathways exploited are (NHEJ) and (HDR), with NHEJ predominating in most cell types due to its efficiency and lack of requirement for a homologous template.01131-X) NHEJ directly ligates DSB ends, often introducing small insertions or deletions (indels) at the junction, which frequently results in frameshift mutations that disrupt function and are harnessed for gene knockouts.00111-9) This pathway operates throughout the , making it suitable for editing in both dividing and quiescent cells, though its error-prone nature limits applications to loss-of-function edits. In contrast, HDR utilizes a homologous donor template to accurately repair DSBs, facilitating precise insertions, deletions, or substitutions and is thus exploited for corrective edits or integration. , including subpathways like synthesis-dependent strand annealing and double resolution, is restricted to S and phases when are available as templates, rendering it less efficient—typically competing with NHEJ at ratios where NHEJ prevails in non-synchronized cells.00111-9) To enhance , strategies such as inhibiting NHEJ factors (e.g., DNA-PK) or synchronizing cells to proliferative phases have been developed, though remains challenging in primary and non-dividing cells. An alternative DSB repair mechanism, microhomology-mediated end joining (MMEJ), serves as a backup pathway involving short homologous sequences (5-25 base pairs) flanking the break for annealing, leading to precise but error-prone joining with deletions of intervening sequences. In genome editing, MMEJ is leveraged in approaches like precise integration into target chromosomes (PITCh) for scarless insertions without long homology arms, particularly useful when HDR is inefficient, though it can also contribute to unintended large deletions. MMEJ activity increases under conditions suppressing classical NHEJ, such as in certain cancer cells deficient in NHEJ components, highlighting its role in editing outcomes influenced by cellular context. These pathways' competition determines editing fidelity, with outcomes varying by locus, cell type, and DSB-end processing factors like end resection, which favors HDR over NHEJ.00200-6)

Principles of Sequence-Specific Targeting

Sequence-specific targeting in genome editing fundamentally relies on engineered nucleases that bind to and cleave DNA at predetermined loci, exploiting endogenous repair pathways for modifications such as insertions, deletions, or substitutions. This specificity is achieved through modular DNA-binding domains that recognize particular nucleotide sequences, typically 12–20 base pairs long, fused to a catalytic nuclease domain that induces double-strand breaks (DSBs). The binding domains operate via direct interactions with DNA bases, ensuring localized nuclease activity while minimizing off-target effects, though imperfect specificity remains a challenge requiring ongoing optimization. In protein-DNA recognition systems, such as those in nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), specificity arises from arrays of protein modules that contact DNA through hydrogen bonds and hydrophobic interactions. Each module in ZFNs typically recognizes a 3–4 subsite, with 3–6 fingers forming a recognition arm of 9–18 bp, though binding affinity is influenced by adjacent fingers and spacer sequences of 5–7 bp between dimerizing monomers. TALENs utilize TALE repeats from plant pathogens, where each 34-amino-acid repeat's repeat-variable di-residues (RVDs) specify a single —e.g., NI for —enabling straightforward programming of longer targets (12–20 bp) with spacers of 12–19 bp. These systems often employ domains, which require dimerization for cleavage, enhancing specificity by necessitating paired binding events. RNA-guided mechanisms, exemplified by CRISPR-Cas systems, achieve targeting through Watson-Crick base pairing between a single-guide RNA (sgRNA) and the target DNA, typically spanning 20 nucleotides adjacent to a protospacer-adjacent motif (PAM) required for Cas nuclease activation, such as NGG for Streptococcus pyogenes Cas9. This RNA-DNA hybridization simplifies programming compared to protein engineering, as only the sgRNA sequence needs alteration, but specificity depends on minimizing mismatches, with off-target cuts occurring at sites with partial complementarity. Unlike protein-based dimers, Cas9 functions as a single polypeptide, scanning DNA for PAMs before R-loop formation and cleavage, though variants like high-fidelity Cas9 mutants reduce unintended activity by altering contact dynamics. Across all approaches, target site accessibility, chromatin state, and cellular repair context influence editing efficiency, underscoring the need for empirical validation of specificity.

Primary Editing Technologies

Meganucleases

Meganucleases, also known as homing endonucleases, are naturally occurring site-specific endonucleases derived primarily from microbial mobile genetic elements, such as introns and inteins. These enzymes recognize extended DNA sequences, typically 12 to 40 base pairs in length, which enables highly precise cleavage with minimal off-target effects due to the rarity of such long motifs in genomes. Unlike modular nucleases, meganucleases integrate DNA-binding and catalytic activities within a single polypeptide, often exhibiting a saddle-shaped structure that cradles the DNA helix. The adaptation of meganucleases for genome editing began in the early , with natural variants like I-SceI from mitochondria used to induce double-strand breaks (DSBs) in mammalian cells as early as 1994, demonstrating enhanced efficiency. Engineered custom meganucleases emerged in the late 1990s and early , pioneered by groups including those at Cellectis, which developed variants through starting around 1999. Redesign strategies, such as semi-rational and in vitro recombination of monomeric domains from dimeric scaffolds like I-CreI (a 22-base-pair recognizer from ), allow tailoring to novel targets, though success rates remain low owing to the coupled evolution of recognition and cleavage domains. In editing applications, meganuclease-induced DSBs trigger cellular repair pathways, including error-prone for gene knockouts or for precise insertions and corrections when donor templates are provided. Their specificity arises from multiple bonds and van der Waals contacts across the target, conferring advantages like reduced and compared to heterologous fusion proteins. However, challenges—such as the need for extensive screening to avoid partial specificities or catalytic inactivity—limit versatility, with redesign often requiring months of iterative optimization. Early applications targeted therapeutic corrections, such as disrupting proviral DNA or correcting mutations in models, and agricultural modifications in plants. Despite these proofs-of-concept, meganucleases have seen limited clinical translation due to design complexity, paving the way for successor technologies like nucleases that offer modular assembly. Ongoing refinements, including machine learning-assisted design, aim to enhance predictability for bespoke nucleases.

Zinc Finger Nucleases

Zinc finger nucleases (ZFNs) are engineered restriction enzymes comprising zinc finger protein domains for sequence-specific DNA recognition fused to the non-specific DNA cleavage domain of the FokI endonuclease. These modular proteins induce targeted double-strand breaks (DSBs) at predetermined genomic loci, exploiting cellular DNA repair pathways such as non-homologous end joining (NHEJ) for gene disruption or homology-directed repair (HDR) for precise insertions or corrections. Each zinc finger module typically binds a 3-base-pair subsite, with arrays of 3–6 fingers providing specificity spanning 9–18 base pairs; FokI dimerization requires two adjacent ZFNs binding in a tail-to-tail orientation, spaced 4–6 base pairs apart, to generate the DSB. Development of ZFNs began with the identification of motifs in the TFIIIA from laevis in 1985, followed by demonstrations of their customizable DNA-binding properties in the early . Pioneering work in the late and early 2000s by researchers including Carlos Barbas and David Liu enabled the fusion of arrays to , achieving the first targeted DSBs in mammalian cells around 2002–2005. Key milestones include the 2009 demonstration of efficient ZFN-mediated editing in human cells via modular , facilitating broader adoption for . Despite early promise as the first programmable genome editing tool, ZFN design proved labor-intensive due to context-dependent interactions between adjacent fingers, often requiring empirical selection or proprietary oligomerized methods like OPEN or . ZFNs have been applied in preclinical models for gene knockouts, insertions, and corrections, notably in disrupting the gene for resistance in human cells and hematopoietic stem cells (HSCs). Clinically, Sangamo Therapeutics advanced ZFN-based therapies, with Phase 1/2 trials for (SB-728) initiating in 2009, showing transient viral load reductions but limited long-term efficacy due to editing efficiency constraints. For hemophilia B, an ZFN approach via AAV delivery (SB-525/ST-920) entered trials in 2018, aiming to insert a transgene into the albumin locus; a 2022 first-in-human study reported safe dosing up to 5×10^13 vg/kg with FIX activity increases, though no approvals have been granted as of 2025. Recent studies in 2024 confirmed high-efficiency ZFN editing in HSCs for multilineage engraftment, underscoring persistent utility in applications. Advantages of ZFNs include proven clinical tolerability, reduced compared to some alternatives, and high specificity when optimized, with off-target effects mitigated by paired design and . However, challenges persist: design complexity limits accessibility, potential toxicity at high expression levels, and off-target cleavage at sites with partial , though rates are generally lower than early iterations when using validated ZFNs. Persistent expression risks promiscuous binding, prompting strategies like mRNA for ephemeral activity. While eclipsed by simpler tools like -Cas9 post-2012, ZFNs remain relevant for applications demanding compact payloads or established safety profiles in viral vectors.

TALENs

Transcription activator-like effector nucleases (TALENs) are engineered restriction enzymes consisting of a customizable derived from transcription activator-like (TAL) effectors of fused to the nonspecific DNA cleavage domain of the FokI endonuclease. TAL effectors, first characterized in 2009, contain tandem repeats with repeat-variable di-residues (RVDs) that confer nucleotide-specific DNA binding, where each RVD typically recognizes a single . The initial demonstration of TALEN-mediated genome editing was reported in 2010, with key publications in 2011 enabling targeted double-strand breaks (DSBs) in various organisms. TALENs function by designing pairs of proteins that bind to adjacent DNA sequences separated by a spacer of 12-20 base pairs; the FokI domains dimerize across this spacer to generate a DSB, which is repaired via (NHEJ) for gene disruption or () for precise edits when a donor template is provided. This modular one-to-one RVD-nucleotide recognition simplifies target design compared to zinc finger nucleases (ZFNs), which rely on modules recognizing three each, often requiring empirical optimization due to context-dependent binding. TALEN assembly, though initially labor-intensive via methods like , has been streamlined with kits allowing construction in 1-2 days. TALENs exhibit higher specificity than ZFNs, with studies showing reduced off-target cleavage at sites like ; for instance, TALENs produced fewer unintended mutations than ZFNs targeting the same locus. Relative to CRISPR-Cas9, TALENs demonstrate lower off-target activity in some contexts due to the absence of mismatches and reliance on protein-DNA interactions, though CRISPR's ease of use has led to its dominance. However, TALENs' larger size (around 3 kb per ) complicates , particularly in vectors, and multiplexing multiple targets remains challenging without custom engineering. Applications of TALENs span basic research and therapeutics, including gene knockouts in human pluripotent stem cells for disease modeling, such as generating mutants resistant to . In agriculture, TALENs have conferred rice resistance to Xanthomonas oryzae by disrupting susceptibility genes and enabled the first genome-edited pigs in 2015 via embryo injection. Therapeutically, TALENs achieved the first cure of in a patient in 2015 by editing T cells for adoptive transfer, highlighting their clinical potential despite subsequent shifts toward . TALENs also facilitate mitochondrial DNA editing for diseases like , exploiting their protein-based delivery to bypass nuclear issues.

CRISPR-Cas Systems

CRISPR-Cas systems derive from clustered regularly interspaced short palindromic repeats () and associated proteins, which form an adaptive immune mechanism in and to defend against invading bacteriophages and plasmids. These systems acquire short DNA sequences from foreign invaders, integrate them as spacers into the host CRISPR array, and transcribe them into CRISPR RNAs (crRNAs) that guide effector proteins to cleave matching nucleic acids during subsequent exposures. The functional role was first demonstrated in 2007 when spacers from phage DNA conferred resistance in . Classified into two main classes, six types, and numerous subtypes, CRISPR-Cas systems vary in complexity and effectors; type II systems, prevalent in genome editing applications, rely on a single large Cas9 endonuclease. In natural type II systems, such as from , Cas9 forms a complex with crRNA and (tracrRNA), which base-pairs with the target DNA to form an structure, enabling double-strand breaks (DSBs) adjacent to a (PAM), typically 5'-NGG-3'. The dual crRNA-tracrRNA was simplified into a single (sgRNA) for programmable targeting. Adaptation for genome editing began with the 2012 demonstration that S. pyogenes Cas9 (SpCas9), guided by dual RNAs, cleaves plasmid DNA in vitro at sites specified by the crRNA spacer sequence, provided a PAM is present. This RNA-guided nuclease activity was harnessed for eukaryotic genome engineering in early 2013, when Cong et al. reported targeted cleavage and homology-directed repair in human and mouse cells using SpCas9 and sgRNA expressed from plasmids, achieving up to 25% modification efficiency at select loci. Independent work by Mali et al. confirmed multiplex editing capabilities, altering up to five endogenous sites simultaneously via NHEJ or HDR pathways. The editing mechanism exploits Cas9-induced DSBs repaired by (NHEJ), often introducing insertions/deletions (indels) for gene disruption, or (HDR) with donor templates for precise insertions or substitutions. Targeting specificity stems from ~20-nucleotide sgRNA-DNA complementarity, though mismatches can reduce efficiency; off-target effects arise from partial hybridization at non-canonical sites with compatible PAMs. SpCas9 requires a 3' PAM, limiting accessible to ~12.5% of the , prompting variants like SpCas9-NG (recognizing 5'-NG-3') or smaller Cas12a (Cpf1) from Francisella novicida, which uses 5'-TTV-3' PAM and generates staggered cuts for scarless . Cas13 variants, such as LwaCas13a, target and cleave single-stranded RNA rather than DNA, enabling transcript knockdown or editing without genomic alterations, though with collateral RNA cleavage upon activation. These systems' simplicity, multiplexing potential, and low cost—relative to protein-based nucleases like ZFNs or TALENs—drove rapid adoption, with over 10,000 publications by 2017 citing CRISPR for editing. Challenges include immunogenicity of bacterial Cas proteins and delivery barriers in vivo, addressed through humanized variants or alternative Cas orthologs.

Base Editing and Prime Editing

Base editing, developed by Alexandrox Komor and colleagues in David Liu's laboratory and first reported in 2016, enables the precise of a target (C) to (T) in DNA without generating double-strand breaks (DSBs). This approach fuses a catalytically impaired protein—either a nickase variant (nCas9) that creates a single-strand nick or a dead Cas9 (dCas9) lacking nuclease activity—with a cytidine deaminase enzyme, such as APOBEC1, to form a cytosine base (CBE). A single-guide RNA (sgRNA) directs the complex to the target site, where the deaminase chemically modifies to uracil (U) within a narrow window of 4-5 nucleotides; during replication or repair, U is recognized as T, resulting in a C·G to T·A . This DSB-free mechanism substantially reduces insertions/deletions (indels) compared to traditional CRISPR- , which relies on error-prone non-homologous end joining (NHEJ), though CBEs can produce bystander edits at adjacent cytosines and exhibit some off-target activity. In 2017, Gaudelli et al. extended base editing to (A) bases with an base editor (ABE), fusing an evolved tRNA (TadA*) to nCas9, enabling programmable A·T to G·C changes via of A to (I), which is templated as G during replication. Subsequent optimizations, including second- and third-generation editors with uracil glycosylase inhibitor (UGI) fusions to suppress pathways that could revert edits, have improved efficiency to over 50% in mammalian cells for many targets while minimizing indels to below 1%. Base editors have demonstrated utility in correcting pathogenic point mutations, such as those in models, but limitations persist, including restricted transition types (only C·G→T·A and A·T→G·C), PAM sequence constraints from , and potential off-targeting from deaminase activity. Prime editing, introduced by Andrew Anzalone and colleagues in David Liu's group in 2019, represents an advanced iteration that permits "search-and-replace" modifications, including all four transition types, small insertions (up to 44 nucleotides), and deletions (up to 80 nucleotides), without DSBs or donor DNA templates. The system employs a prime editor protein—a fusion of nCas9 and a Moloney murine leukemia virus reverse transcriptase (M-MLV RT)—guided by a prime editing guide RNA (pegRNA) that extends beyond standard sgRNAs to include a reverse transcriptase template (RTT) specifying the desired edit and a primer binding site (PBS) for reverse transcription initiation. Upon binding, nCas9 nicks the target strand (typically the non-template strand), the exposed 3' flap hybridizes to the PBS, and RT copies the RTT into a new DNA flap, which ligases into the genome after flap resolution, displacing the original sequence. Initial efficiencies reached 20-50% for transitions in human cells with minimal indels (<1-5%), outperforming homology-directed repair (HDR) in non-dividing cells, though prime editing historically suffers from lower yields for insertions/deletions and sensitivity to pegRNA design. Engineered variants, such as PE2 with an improved RT and PE3 incorporating an additional sgRNA for nicking the non-edited strand to bias repair, have boosted efficiencies up to 2.3-fold, while recent ePE and ProPE systems further expand the editing window and reduce byproducts. Prime editing's versatility addresses base editing's limitations by enabling transversions indirectly via multi-step edits or hybrid approaches, with off-target rates comparable to or lower than Cas9 due to the requirement for precise RT priming. However, challenges include pegRNA production complexity, potential cellular toxicity from RT activity, and efficiencies still lagging behind DSB-based methods for some large edits, prompting ongoing refinements like smaller Cas variants for delivery. Both technologies, recognized with the 2025 Breakthrough Prize in Life Sciences awarded to David Liu, exemplify a shift toward DSB-independent editing to enhance safety and precision in therapeutic contexts.

Novel and Hybrid Approaches

Novel approaches in genome editing extend beyond conventional nuclease-based systems by incorporating elements such as transposases, integrases, and retrons to enable precise insertions, reductions in double-strand breaks (DSBs), and multiplexing capabilities. Hybrid systems, which fuse components with other molecular machinery, aim to mitigate off-target effects and DSB-associated risks like indels or chromosomal rearrangements, while facilitating large payload integrations up to several kilobases. These innovations, emerging prominently since the early 2020s, prioritize DNA repair-independent mechanisms to enhance efficiency in therapeutic and research applications. CRISPR-associated transposases (CASTs) represent a hybrid class that couples type I CRISPR RNA-guided targeting with transposase activity for programmable DNA insertion. Unlike DSB-dependent methods, CASTs catalyze strand transfer to insert payloads without breaks, achieving efficiencies up to 40% in bacterial systems and demonstrating adaptability to eukaryotic cells through laboratory evolution. For instance, evoCAST variants, optimized via directed evolution, have enabled precise integrations in human cell lines with minimal off-target activity. These systems, identified in diverse prokaryotes, bypass homology-directed repair limitations and support cargo sizes exceeding 10 kb, positioning them for applications in gene therapy where stable, large-scale modifications are required. Programmable addition via site-specific targeting elements (PASTE) exemplifies a hybrid nuclease-integrase fusion, employing a CRISPR-Cas9 nickase linked to a reverse transcriptase and serine integrase for DSB-free large-sequence insertions. Developed in 2022, PASTE uses prime editing-inspired pegRNA to prime reverse transcription of donor DNA, followed by integrase-mediated attachment at nicked sites, yielding up to 25% efficiency for 36 kb inserts in human cells. This approach excels in replacing entire defective genes, such as modeling Duchenne muscular dystrophy by inserting micro-dystrophin cassettes, and avoids DSB toxicity, though delivery challenges persist for in vivo use. Retron-based editing leverages bacterial retrons—RNA-templated reverse transcriptases producing multi-copy single-stranded DNA (ssDNA)—as donor templates for precise homology-directed repairs, often hybridized with for targeting. In a 2025 advancement, retron systems corrected large disease-related mutations in vertebrate models by excising defective regions and inserting healthy sequences, achieving higher fidelity than traditional donors due to in situ ssDNA generation. Efficiencies reach over 50% in mammalian cells when paired with , with retrons enabling multiplex edits via parallel msDNA production, though optimization for payload size and host compatibility continues. This method's repair independence from cell cycle phase broadens its utility across kingdoms. Multiplex automated genome engineering (MAGE), a non-nuclease hybrid relying on recombineering with short ssDNA oligos and phage-derived recombinases, facilitates simultaneous edits at hundreds of loci in prokaryotes. Introduced around 2009 and refined for eukaryotes, MAGE cycles oligonucleotide electroporation with selection to evolve genomes rapidly, as seen in recoding E. coli with over 300 changes for non-canonical amino acid incorporation. While less reliant on sequence-specific nucleases, its integration with CRISPR hybrids enhances scalability for synthetic biology, though eukaryotic efficiencies lag at under 10% per site without further engineering.

Delivery Systems and Implementation Strategies

Viral and Non-Viral Delivery Methods

Viral vectors leverage the natural infectivity of viruses to deliver genome editing components, such as and , into target cells with high efficiency. Adeno-associated viruses (AAVs), particularly serotypes like AAV2 and AAV9, are favored for their non-pathogenic nature, ability to transduce post-mitotic cells, and episomal persistence without genomic integration, supporting transient or long-term expression depending on the application. AAVs have a packaging limit of about 4.7-5 kb, restricting delivery to compact editing systems like or base editors, and have been used in over 150 clinical trials for by 2023, including the FDA-approved in 2017 for RPE65-mediated retinal dystrophy via subretinal AAV delivery achieving sustained vision improvement. However, AAVs can elicit pre-existing neutralizing antibodies in up to 50-70% of humans, potentially reducing efficacy, and high doses may trigger innate immune responses or hepatotoxicity, as observed in a 2020 tragic trial outcome involving AAV for . Lentiviral vectors, derived from , provide larger cargo capacity (up to 9 kb) and integrate into the host genome for stable expression, making them suitable for ex vivo editing of ; they underpinned the FDA approval of in 2017 for leukemia via . Drawbacks include risks of insertional oncogenesis, evidenced by rare leukemia cases in early , and production scalability issues despite advances in integrase-defective variants that promote non-integrating episomal delivery to mitigate genotoxicity. Adenoviral vectors offer high transient expression and larger payloads but provoke strong inflammatory responses, limiting their use to short-term editing in non-immunoprivileged tissues. Non-viral delivery systems circumvent viral immunogenicity and integration risks by employing synthetic or physical carriers for editing components, often as mRNA, plasmids, or ribonucleoproteins (RNPs) to enable transient activity that curtails prolonged off-target editing. Lipid nanoparticles (LNPs), composed of ionizable lipids, cholesterol, and PEG-lipids, encapsulate Cas9 mRNA and guide RNA for systemic in vivo delivery, achieving biodegradability and endosomal escape; they facilitated the first in vivo human CRISPR trial in 2021 for transthyretin amyloidosis (NTLA-2001), with a single dose yielding up to 96% serum protein reduction at 87% liver editing efficiency in phase 1 data reported in 2023. LNPs excel in scalability—billions of doses produced for COVID-19 mRNA vaccines by 2021—and lower mutagenesis risk, but suffer from hepatic tropism, transient expression (hours to days), and potential lipid toxicity at high doses, with editing efficiencies often below 50% in extrahepatic tissues without targeting ligands. Polymer-based nanoparticles, such as polyethyleneimine (PEI) or poly(lactic-co-glycolic acid) (PLGA), offer customizable surface modifications for tissue specificity and protection against nuclease degradation, demonstrating 30-70% editing in mouse glioblastoma models via intracranial injection in 2021 studies. Physical methods like electroporation apply electric pulses to transiently permeabilize membranes, achieving high ex vivo efficiencies (up to 90%) in hard-to-transfect cells like primary T lymphocytes or stem cells without chemical additives, as in Casgevy (exagamglogene autotemcel) approved in 2023 for sickle cell disease following electroporation-mediated BCL11A editing. Yet, electroporation induces cytotoxicity (10-30% cell death) and is impractical for in vivo use due to tissue damage, while hydrodynamic injection or ultrasound-mediated delivery remains experimental with variable yields. Microinjection and nucleofection variants enhance precision in embryos or organoids but scale poorly for therapeutics.
Delivery TypeKey AdvantagesKey DisadvantagesTypical Applications
Viral (e.g., AAV, Lentiviral)High transduction efficiency (50-90% in vivo); natural tropism for tissues like liver, retina, CNSImmunogenicity; limited cargo size (AAV); insertional risks (lentiviral); manufacturing complexityIn vivo therapeutics (e.g., ocular, hepatic editing); ex vivo stem cell modification
Non-Viral (e.g., LNPs, Electroporation)Reduced immunogenicity; transient expression minimizing off-targets; scalable production; no replication riskLower efficiency (10-70%); poor in vivo targeting without modifications; potential cytotoxicityEx vivo cell therapies (e.g., ); emerging in vivo mRNA/RNP delivery for systemic diseases
Hybrid approaches, such as virus-like particles (VLPs) pseudotyped with viral envelopes but lacking genomes, combine viral entry efficiency with non-viral safety, delivering Cas9 RNPs with up to 40% editing in hepatocytes in 2024 preclinical models, though clinical translation lags due to optimization needs. Overall, viral methods dominate current clinical pipelines for their reliability, while non-viral innovations, propelled by mRNA vaccine successes, are advancing toward parity in efficiency and specificity, particularly for transient editing to enhance safety in therapeutic genome editing.

Ex Vivo versus In Vivo Applications

Ex vivo genome editing involves extracting cells from a patient, modifying their genomes in a controlled laboratory environment using tools such as , and subsequently reintroducing the edited cells into the patient. This approach allows for precise manipulation under optimized conditions, including electroporation or viral transduction for delivery, followed by selection or expansion of successfully edited cells to achieve high purity before transplantation. It is particularly suited for accessible cell types like (HSPCs) or T cells, enabling applications in blood disorders and immunotherapies. A primary advantage of ex vivo editing is the ability to mitigate off-target effects and immune responses by editing in isolation, with post-editing validation and enrichment ensuring only viable, correctly modified cells are used. For instance, in the treatment of sickle cell disease (SCD) and transfusion-dependent beta-thalassemia, ex vivo CRISPR-Cas9 editing of patient-derived HSPCs to disrupt the BCL11A enhancer has led to approved therapies like Casgevy (exagamglogene autotemcel), authorized by the FDA in December 2023, demonstrating durable fetal hemoglobin induction and symptom amelioration in clinical trials with over 90% reduction in vaso-occlusive crises. However, challenges include scalability of manufacturing, engraftment efficiency post-infusion, and limitation to ex vivo-accessible tissues, restricting broader use for non-hematopoietic conditions. In contrast, in vivo genome editing delivers editing components—typically via lipid nanoparticles, adeno-associated virus (AAV) vectors, or other systemic/local methods—directly into the patient's body to target cells in situ. This method holds potential for treating organs like the liver, retina, or muscle, where cell extraction is impractical, by achieving site-specific modifications without surgical intervention. Delivery innovations, such as liver-tropic AAVs or nanoparticle-encapsulated Cas9 ribonucleoproteins, have enabled transient expression to reduce prolonged off-target risks. Key benefits of in vivo approaches include broader tissue applicability and avoidance of ex vivo processing complexities, as evidenced by NTLA-2001, an in vivo therapy targeting the TTR gene in hereditary transthyretin amyloidosis (), which achieved up to 87% serum TTR reduction in phase 1 trials via intravenous lipid nanoparticle delivery as of June 2021. Early trials for Leber congenital amaurosis type 10 () have also used subretinal AAV-CRISPR injections to edit CEP290 mutations, restoring partial visual function in preclinical models and initial human dosing by 2020. Nonetheless, hurdles persist, including inefficient targeting of non-dividing cells, potential immunogenicity of bacterial-derived Cas proteins, and amplified safety concerns from systemic distribution, with most trials still in early phases compared to ex vivo successes. Ongoing refinements in delivery specificity aim to bridge these gaps for scalable clinical translation.
AspectEx VivoIn Vivo
Primary ApplicationsHematologic disorders (e.g., , beta-thalassemia), T-cell therapies for cancerLiver diseases (e.g., ), ocular disorders (e.g., ), neuromuscular conditions
Delivery MethodsElectroporation, lentiviral/retroviral vectors in vitro, lipid nanoparticles, direct injection
AdvantagesHigh editing purity via selection; controlled environment reduces immunogenicityTargets inaccessible tissues; no cell extraction needed
ChallengesLimited to harvestable cells; manufacturing and engraftment variabilityDelivery efficiency; off-target effects in vivo; immune clearance of editors
Clinical Status (as of 2025)Multiple approvals (e.g., , 2023); dozens of trialsPhase 1/2 trials dominant (e.g., , 2021 onward); no approvals yet

Technical Performance Metrics

Precision, Efficiency, and Off-Target Effects

Precision in genome editing refers to the accuracy with which a nuclease targets the intended DNA sequence, minimizing unintended modifications elsewhere in the genome. Early tools like exhibit high specificity due to their large recognition domains spanning 12-40 base pairs, but their engineering is labor-intensive, limiting widespread use. Zinc finger nucleases () and transcription activator-like effector nucleases () achieve greater specificity than initial CRISPR systems through modular protein-DNA interactions—ZFNs recognizing 9-18 base pairs via zinc finger arrays and TALENs targeting longer stretches with TALE repeats—resulting in off-target mutation rates often below 0.1% in optimized designs, though efficiencies typically range from 10-50% in mammalian cells due to delivery and repair pathway dependencies. CRISPR-Cas9 systems revolutionized efficiency, enabling editing rates exceeding 80% in many cell types via simple guide RNA (gRNA) design, but early implementations showed off-target cleavage frequencies up to 93.6% at mismatched sites due to tolerance for 1-5 nucleotide mismatches and PAM-adjacent positioning. GUIDE-seq and similar assays revealed off-target rates of 0.1-5% genome-wide in human cells, varying by gRNA sequence and Cas9 variant, with double-strand breaks (DSBs) at non-target sites triggering indels or rearrangements via non-homologous end joining (NHEJ). High-fidelity Cas9 mutants (e.g., SpCas9-HF1, eSpCas9) and paired nickases reduce these by 10- to 100-fold through enhanced gRNA-DNA hybridization stringency, though empirical validation remains essential as in silico predictions underestimate effects by up to 50%. Efficiency metrics, often measured as indel formation or homology-directed repair (HDR) yield, favor CRISPR over ZFNs and TALENs—e.g., CRISPR achieving 2-10x higher on-target editing in HEK293 cells—but HDR remains low (1-20%) across platforms due to NHEJ dominance in dividing cells.00111-9) Base editors and prime editors enhance precision by enabling single-base conversions or small indels without DSBs: cytosine base editors (CBEs) and adenine base editors (ABEs) convert C·G to T·A or A·T to G·C with off-target rates under 0.5%, while prime editors install diverse edits (insertions up to 44 bp, deletions) at 20-50% efficiency, minimizing bystander edits and genomic instability compared to DSB-based methods. These advances, validated in cellular and animal models, underscore causal links between nuclease architecture, mismatch tolerance, and repair outcomes, with ongoing quantification via unbiased sequencing essential for therapeutic viability.

Strategies for Enhancing Specificity and Yield

One prominent strategy for improving specificity involves engineering high-fidelity variants of the Cas9 nuclease to reduce non-specific DNA binding. For instance, SpCas9-HF1 incorporates alanine substitutions at residues N497, R661, Q695, and Q926, which disrupt extraneous interactions with the phosphate backbone, leading to undetectable off-target cleavage events via GUIDE-seq for six out of seven tested single-guide RNAs (sgRNAs) in human cells, while preserving greater than 85% of wild-type on-target activity across 32 sgRNAs. Similarly, the Sniper2L variant, derived from directed evolution of predecessor Sniper-Cas9 with an E1007L mutation in the RuvC domain, maintains on-target indel frequencies comparable to wild-type SpCas9 (up to 50% in HEK293T cells) but exhibits markedly lower off-target indel rates across over 7,000 NGG PAM targets, quantified by a specificity metric of 1 minus the ratio of off-target to on-target frequencies. Additional molecular optimizations enhance specificity by altering guide RNA architecture or nuclease deployment. Truncated sgRNAs (12-18 nucleotides in the spacer) limit the editing window and mismatch tolerance, reducing off-target effects by up to 5-fold in some assays without substantially compromising on-target efficiency. Paired Cas9 nickases, utilizing dual sgRNAs to generate staggered single-strand nicks offset by 4-100 base pairs, further enforce dual-site recognition, achieving near-complete elimination of off-target double-strand breaks compared to wild-type Cas9 in human cell lines.14619-1) Protocol-level adjustments, such as transient delivery of Cas9 ribonucleoproteins (RNPs) at low concentrations or during specific cell cycle phases, temporally restrict nuclease activity to minimize prolonged exposure and bystander cuts. To boost editing yield, particularly for homology-directed repair (HDR) pathways that enable precise insertions or substitutions but occur at rates below 10% in dividing cells due to competition from error-prone non-homologous end joining (NHEJ), strategies target repair pathway bias. Chemical inhibition of NHEJ factors, such as ligation with SCR7 (which blocks DNA ligase IV), combined with HDR-promoting agents like RS-1 (an RAD51 stimulator), has increased HDR efficiency 2- to 5-fold in HEK293T and K562 cells for targeted knock-ins. Cell cycle synchronization via nocodazole arrest in G2/M phase, where HDR machinery peaks, yields up to 3-fold HDR gains, as endogenous repair favors template-directed synthesis during mitosis preparation. Donor template optimizations, including single-stranded oligodeoxynucleotides (ssODNs) positioned within 20-100 base pairs of the cut site, further elevate yields by facilitating invasion of the Cas9-generated overhang. Post-editing enrichment methods amplify yield by selecting edited subpopulations without relying solely on intrinsic efficiencies. HDR surrogate reporters, featuring a frameshifted coding sequence (e.g., BFP to GFP conversion) restored only via precise template integration, enable fluorescence-activated cell sorting to enrich knock-in events by 20.7-fold (from 2.22% to 45.93%) in human cell lines. NHEJ-based reporters similarly select for nuclease-proficient cells, achieving up to 39-fold increases in indel frequencies for genes like TP53. Modular ssDNA donors paired with NHEJ inhibitors or HDR enhancers have reported HDR rates exceeding 90% in optimized systems, though scalability to primary cells remains context-dependent due to variable repair dynamics. These approaches, while effective, often involve trade-offs, such as potential toxicity from inhibitors or reduced multiplexing capacity.

Practical Applications

Therapeutic Uses in Human Medicine

Genome editing technologies, particularly CRISPR-Cas9, have enabled targeted correction of disease-causing mutations in human cells for therapeutic purposes, focusing initially on monogenic disorders amenable to ex vivo modification of accessible cell types like hematopoietic stem cells (HSCs). The pioneering approval of exagamglogene autotemcel (Casgevy) by the U.S. Food and Drug Administration on December 8, 2023, marked the first clinical use of CRISPR-based editing for sickle cell disease (SCD) and transfusion-dependent β-thalassemia (TDT). This therapy involves extracting patient HSCs, using CRISPR-Cas9 to disrupt the BCL11A enhancer to reactivate fetal hemoglobin (HbF) expression, and reinfusing the edited cells after myeloablative conditioning. In phase 1/2 trials, 29 of 31 SCD patients remained free of severe vaso-occlusive crises for at least 12 months post-infusion, with HbF levels reaching 30-40% in most cases, demonstrating durable engraftment and clinical benefit. Beyond hemoglobinopathies, ex vivo editing targets other blood-related conditions and immunotherapies. CRISPR-edited allogeneic CAR-T cells are in trials for cancers like multiple myeloma and B-cell malignancies, aiming to enhance persistence by knocking out PD-1 or TCR genes to reduce exhaustion and graft-versus-host risks. For primary immunodeficiencies such as severe combined immunodeficiency (SCID), trials edit autologous HSCs to restore functional IL2RG expression, with early data showing immune reconstitution in X-SCID patients. These approaches leverage the accessibility of blood cells but require chemotherapy preconditioning, limiting applicability to non-hematopoietic tissues. In vivo editing, delivering nucleases directly to target organs via lipid nanoparticles or AAV vectors, addresses systemic diseases but faces challenges in efficiency and off-target risks. Intellia's NTLA-2001, targeting TTR mutations in hereditary transthyretin amyloidosis (ATTR), achieved up to 87% serum TTR reduction in phase 1/2 trials with a single dose, with phase 3 data expected by late 2025 supporting potential approval for cardiomyopathy and polyneuropathy. For Leber congenital amaurosis type 10, Editas Medicine's EDIT-101 (AAV-delivered CRISPR) improved visual acuity in 79% of treated eyes in the phase 1/2 BRILLIANCE trial, though long-term efficacy remains under evaluation. Emerging pipelines explore in vivo applications for muscular dystrophy (e.g., editing DMD exons), cystic fibrosis (CFTR correction in lung epithelium), and HIV (CCR5 disruption in CD4+ T cells), with over 150 active CRISPR trials as of mid-2025 spanning these areas, though most remain in phases 1-2 due to delivery and immunogenicity hurdles. Base and prime editing variants promise higher precision for point mutations, with preclinical advances in trials for SCD alternatives and alpha-1 antitrypsin deficiency. Therapeutic editing's success hinges on disease biology favoring high editing rates (e.g., >20% allelic correction in HSCs for HbF induction) and tolerable safety profiles, with adverse events primarily from conditioning rather than editing itself in approved cases. Ongoing challenges include scalability for rare diseases and equitable access, as Casgevy's list price exceeds $2 million per treatment. No additional genome editing therapies have gained regulatory approval by October 2025, underscoring the field's nascent stage despite rapid trial expansion.

Agricultural and Plant Breeding Improvements

Genome editing has revolutionized plant breeding by enabling precise modifications to endogenous genes, bypassing the lengthy cycles of conventional cross-breeding and reducing reliance on random mutagenesis. Technologies like CRISPR-Cas9 allow for targeted knockouts, insertions, or base edits that introduce traits such as enhanced yield or stress tolerance without incorporating foreign DNA, distinguishing them from transgenic GMOs. By 2024, over 25 plant species had been successfully edited across more than 100 genes, yielding improvements in agronomic performance. In disease resistance, CRISPR-mediated editing has targeted susceptibility genes to confer broad-spectrum protection. For instance, in wheat, multiplexed editing of TaMLO genes produced varieties resistant to powdery mildew, a fungal pathogen causing significant yield losses, with edited lines showing up to 90% reduced infection rates under field conditions. Similar approaches in rice have disrupted SWEET genes to enhance resistance to bacterial blight, while in bananas, editing of susceptibility loci has improved tolerance to Fusarium wilt, a devastating soil-borne disease threatening global production. These modifications demonstrate causal links between gene disruption and pathogen evasion, validated through controlled inoculation trials. For and tolerance, editing phytohormone pathways has shortened plant stature in via gibberellin-related mutants, reducing and boosting without yield penalties. In and soybeans, CRISPR edits to flowering-time genes have accelerated maturity, enabling adaptation to shorter growing seasons amid climate variability, with field trials reporting 10-20% increases under . Nutritional enhancements include elevating β-carotene levels sixfold in and bananas through PSY gene activation, addressing deficiencies in staple crops consumed by billions. Regulatory frameworks influence deployment: the U.S. USDA exempts certain site-directed nuclease-1 (SDN-1) edits—those mimicking natural mutations—from oversight if no plant pest risks are introduced, as revised in regulations, enabling commercialization of products like herbicide-tolerant canola by 2023. In contrast, the EU's precautionary approach classifies most edited plants as GMOs under Directive 2001/18/EC, imposing rigorous assessments despite absent transgenes, though a 2023 proposal seeks to deregulate "NGT-1" plants akin to conventional varieties. This disparity highlights how evidence-based risk assessments in the U.S. have accelerated adoption, with over a dozen edited crops entering markets by 2024, versus stalled EU pipelines. Empirical data from multi-year field studies underscore the safety and efficacy of these edits, with no verified off-target effects leading to unintended traits in commercial lines.

Animal Models and Basic Research

Genome editing technologies, particularly , have enabled the rapid generation of genetically modified animal models that recapitulate human disease phenotypes and elucidate gene functions . Unlike earlier methods such as zinc-finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs), which required complex , systems use guide RNAs for programmable targeting, achieving higher efficiency in injection and modification across . This has accelerated by allowing multiplexed knockouts—simultaneous disruption of multiple genes—to study genetic interactions and pathways, as demonstrated in models where achieved editing efficiencies exceeding 80% in founder animals. In , CRISPR-Cas9 has been pivotal for modeling monogenic and polygenic diseases. For instance, targeted knockouts in mice have revealed causal roles of specific genes in neurodegeneration, such as Apolipoprotein E variants in pathology, with models exhibiting amyloid plaque accumulation and tau hyperphosphorylation mirroring human findings. Rat models, optimized via of Cas9 protein and single-guide RNAs into zygotes, have improved behavioral assays for psychiatric disorders, yielding rates up to 100% for single loci without off-target indels in coding regions. These models surpass traditional transgenic approaches in speed, with full germline transmission achievable in one generation, facilitating through loss-of-function studies. Zebrafish and Xenopus serve as high-throughput platforms for developmental and metabolic research due to their optical transparency and , enabling real-time imaging of edited embryos. CRISPR-induced mutations in have modeled skeletal dysplasias, such as those from Col2a1 disruptions, uncovering mechanisms of formation absent in systems. Base editing variants, which introduce precise single-nucleotide changes without double-strand breaks, have expanded this to models of metabolic syndromes, achieving correction efficiencies of 20-50% and reducing mosaicism compared to standard . In larger mammals like pigs, TALENs and have generated models for cardiovascular diseases by knocking out , resulting in phenotypes validated against lipid profiles. These animal models underscore genome editing's utility in dissecting causal gene-environment interactions, though limitations persist: species-specific differences in DNA repair pathways can alter editing outcomes, and off-target effects, though minimized to below 1% with high-fidelity variants, necessitate rigorous validation via whole-genome sequencing. Ongoing refinements, including for scarless insertions, promise even greater fidelity for into complex traits.

Industrial Biotechnology

In industrial biotechnology, genome editing technologies such as CRISPR-Cas9 have enabled precise modifications to microbial genomes, optimizing metabolic pathways for the scalable production of biofuels, biochemicals, enzymes, and amino acids. These tools facilitate targeted knockouts, insertions, and promoter optimizations in organisms like Escherichia coli, Saccharomyces cerevisiae, and Clostridium species, which are commonly used in fermentation processes. By altering genes involved in substrate utilization, byproduct inhibition, and product flux, editing enhances yields and process efficiency, reducing reliance on chemical synthesis and fossil fuels. For biofuel production, CRISPR-Cas9 has been applied to improve solventogenesis in anaerobic bacteria. In Clostridium beijerinckii, editing alcohol dehydrogenase genes (adhE1 and adhE2) increased n-butanol titers while maintaining strain stability, addressing acid buildup that hampers industrial scalability. Similarly, in Clostridium acetobutylicum, disruption of acid formation pathways shifted metabolism toward butanol, elevating the butanol-to-acetate ratio and supporting higher solvent yields. In yeast, overexpression of lipid elongation genes via CRISPR boosted ethanol productivity, with reported improvements in S. cerevisiae strains for bioethanol fermentation as of 2021. Chemical production has seen notable gains through microbial chassis engineering. CRISPR-mediated deletions of eight genes in S. cerevisiae amplified fatty acid output by 30-fold, enabling efficient conversion of glucose to lipids for bioderived fuels or materials. In Klebsiella pneumoniae, pathway optimizations yielded a ~50% increase in 1,3-propanediol, a precursor for polymers, demonstrated in 2022 studies. For E. coli, editing lactate dehydrogenase (ldhA), alcohol dehydrogenase (adhE), and related genes raised succinate production to 80 g/L, a key dicarboxylic acid for bioplastics. Bacterial strains engineered for squalene from glucose achieved high-efficiency terpenoid synthesis by 2019. Enzyme and amino acid biosynthesis benefit from cofactor balancing and secretion enhancements. In Corynebacterium glutamicum, CRISPR targeting NADPH supply genes and promoters improved L-glutamate titers for feed and pharmaceuticals, with advancements reported in 2021. Bacillus subtilis strains edited at the PrsA chaperone locus exhibited enhanced amylase secretion in 2024, streamlining industrial enzyme production for detergents and biofuels. Tryptophan yields in E. coli rose ~40% via enzyme pathway tweaks in 2019, while Aspergillus niger optimizations for citric acid fermentation improved efficiency by 2019. Companies like Ginkgo Bioworks leverage these techniques for custom microbial factories producing pharmaceuticals and fine chemicals.

Clinical Translation and Regulatory Milestones

Landmark Trials and Approved Therapies

The first approved genome editing therapy utilizing CRISPR-Cas9, Casgevy (exagamglogene autotemcel), received U.S. (FDA) approval on December 8, 2023, for (SCD) in patients aged 12 years and older with recurrent vaso-occlusive crises requiring annual intravenous infusions. Developed jointly by and , Casgevy employs editing to disrupt the BCL11A enhancer in hematopoietic cells, thereby reactivating production to mitigate SCD's pathological effects. Approval for transfusion-dependent beta-thalassemia (TDT) followed on January 16, 2024, based on phase 1/2/3 trials (CLIMB-121 for SCD and CLIMB-131 for TDT) demonstrating 96.6% of SCD patients free from severe vaso-occlusive crises at 12 months post-infusion and 93.5% of TDT patients achieving transfusion independence at the same interval, though outcomes required myeloablative preconditioning with associated risks including and secondary malignancies. Preceding Casgevy's approval, the inaugural human application of CRISPR occurred in October 2016 at in , where three patients with advanced received autologous T cells edited via -Cas9 to knock out the PD-1 gene, aiming to enhance antitumor immunity; phase 1 data reported in 2020 confirmed feasibility and absence of severe off-target effects or oncogenic insertions, though clinical responses were modest with no complete remissions observed. In the United States, the first CRISPR trial commenced in 2019 under leadership, infusing patients with refractory cancers (, , ) with T cells edited to express a NY-ESO-1-specific alongside knockouts of PD-1 and TRAC loci; interim phase 1 results indicated durable of edited cells and objective responses in some participants, establishing safety for multiplex editing without unanticipated . A pivotal advancement in genome editing materialized in March 2020 at (OHSU), marking the first direct intraocular injection of CRISPR-Cas9 components (via AAV5 vector) to treat caused by CEP290 mutations; the phase 1/2 trial (NCT03872479) targeted the IVS26 intronic variant, with initial safety data from the first cohort showing no serious adverse events and modest improvements in some eyes at 6 months, though efficacy varied and long-term immunogenicity risks persisted. Complementing successes, and Regeneron's NTLA-2001 trial in 2021 represented the first systemic CRISPR application for transthyretin , achieving up to 87% serum protein reduction at single 0.3 mg/kg doses in phase 1, with durable effects through 24 months and no dose-limiting toxicities, underscoring CRISPR's potential for liver-directed editing without cell extraction. By 2025, Casgevy remained the sole fully approved CRISPR-based therapy globally, with conditional authorizations from the for TDT in December 2023 and SCD thereafter, alongside FDA approval in 2024 for both indications in s 12 and older; no additional approvals had materialized despite over 250 ongoing trials, many targeting and autoimmune disorders via allogeneic edited cells like ' CTX112. A notable 2025 milestone involved the first personalized therapy administered to a pediatric with a rare at and Penn Medicine, using base editing to correct a unique patient-specific ex vivo, demonstrating rapid metabolic normalization without evident toxicity in this single-case intervention reported in May. These developments highlight empirical progress in safety and targeted efficacy, tempered by challenges in scalability, preconditioning toxicities, and off-target editing verification across diverse genetic backgrounds.

Global Research Pipelines and Challenges

Global research pipelines in genome , particularly CRISPR-based therapies, have expanded rapidly, with approximately 250 clinical trials registered worldwide as of February 2025, of which over 150 remain active, primarily targeting blood disorders, cancers, and inherited conditions. Leading efforts are concentrated , where companies like advance programs such as CTX310 (Phase 1 trial initiated for via ANGPTL3 targeting, with data expected in late 2025) and CTX320 (ongoing Phase 1 for ). and , pipelines focus on applications, including Editas Medicine's use of and Cas12a nucleases for ocular and blood disorders, while Chinese trials emphasize scalable manufacturing for hemoglobinopathies. By late 2024, over 50 active treatment centers operated globally, spanning at least 46 countries, though the U.S. hosts the majority of advanced-stage trials. Broader gene efforts contribute to nearly 3,500 preclinical and clinical programs in cell and gene therapies, reflecting a shift toward multiplex and for complex diseases like and . Key pipelines integrate diverse editing modalities, with pursuing immuno-oncology and alongside approved ex vivo therapies like Casgevy for . Emerging trials, such as those targeting disorders, demonstrate progress in non-viral delivery systems, though most remain in early phases (Phase 1/2). International collaborations, including those under the Innovative Genomics Institute, aim to standardize protocols, but pipelines vary by region: U.S.-led efforts prioritize FDA-aligned safety data, while China's (NMPA) has approved faster tracks for certain ZFNs and applications in . Challenges in clinical translation persist across technical, regulatory, and logistical domains. Off-target edits and structural variations, including unintended deletions of regulatory elements, pose genotoxic risks that demand enhanced specificity assays beyond standard sequencing. Delivery remains a bottleneck, with viral vectors facing immune responses and scalability issues, while non-viral alternatives like lipid nanoparticles yield lower efficiencies . Regulatory hurdles exacerbate delays: disparate frameworks between the FDA, , and NMPA complicate multinational trials, with unclear guidelines on (GMP) reagents and long-term biodistribution data hindering approvals. Manufacturing scalability and cost represent additional barriers, as personalized ex vivo editing inflates expenses—often exceeding $2 million per patient—limiting access despite curative potential in trials for blood cancers. Ethical and safety considerations, including non-clinical requirements for germline off-target scrutiny even in somatic applications, slow progression, with calls for platform-based regulatory solutions to harmonize global standards. Despite these, empirical successes in Phase 3 trials underscore feasibility, though unresolved uncertainties in durable editing efficacy necessitate rigorous, multi-year follow-up.

Risks and Empirical Limitations

Biological Hazards and Failure Modes

Genome editing via induces double-strand breaks that are primarily repaired by error-prone (NHEJ), leading to insertions or deletions (indels) that frequently cause frameshift mutations and gene disruption. Off-target effects arise from cleaving unintended sites due to sequence mismatches tolerated up to three base pairs, resulting in unintended mutations at frequencies as low as 0.03% but potentially higher depending on design. Mosaicism occurs when editing efficiency varies across cells, particularly in embryos, producing heterogeneous populations with mixed edited and unedited genotypes, complicating therapeutic outcomes and transmission. On-target aberrations include large-scale structural variations such as kilobase- to megabase-scale deletions, chromosomal truncations, and , often undetected by short-read sequencing. Chromosomal translocations between homologous or sites can form unstable dicentric or acentric chromosomes, with frequencies amplified up to 1000-fold by certain inhibitors. In clinical contexts, such as BCL11A editing in exa-cel therapy for , kilobase deletions have been observed, potentially disrupting . Biological hazards encompass immune responses against proteins, particularly in patients with pre-existing antibodies, which can destroy edited cells and undermine efficacy applications. These failure modes heighten oncogenic risks if edits affect proto-oncogenes or tumor suppressors, as unintended structural changes or indels may promote genomic instability and tumorigenesis. Large DNA insertions or deletions induced by have been empirically linked to increased cancer risk in cell models. Overall, while mitigation strategies like high-fidelity variants reduce off-target rates by up to 98.7%, persistent uncertainties in long-read detection underscore the need for comprehensive safety assessments.

Long-Term Safety Data and Uncertainties

As of 2025, long-term safety data for genome editing technologies, particularly CRISPR-Cas systems, remains limited due to their relatively recent development and deployment, with most human applications initiated post-2018. Animal studies provide the primary longitudinal evidence; for instance, in mouse models of retinal editing, CRISPR-Cas9 modifications demonstrated sustained efficacy and low off-target mutation rates (1.28% at 196 days post-injection, rising modestly to 2.27% at 585 days), with no overt toxicity observed over nearly two years. Similarly, non-human primate trials for conditions like hemophilia have shown stable edits persisting for over a year without systemic adverse effects, though sample sizes are small and follow-up durations rarely exceed three years. These findings suggest short- to medium-term stability in controlled somatic applications, but extrapolation to humans requires caution given species-specific differences in DNA repair and immune responses. In clinical contexts, approved therapies like exagamglogene autotemcel (Casgevy) for and beta-thalassemia, authorized by the FDA in December 2023, have reported no off-target-related adverse events in initial follow-ups of up to two years, with patients achieving durable corrections. However, these datasets derive from small cohorts (e.g., 44 patients in pivotal trials), and long-term monitoring—essential for detecting delayed risks such as oncogenesis from unrepaired double-strand breaks—is ongoing but incomplete, with median follow-up under three years as of mid-2025. Peer-reviewed analyses of early trials emphasize that while base editing and variants reduce off-target frequencies to below 1% , real-world genomic instability, including large deletions or translocations, has been documented in up to 5% of edited lines. Key uncertainties stem from potential cumulative effects of off-target edits, which could induce chromosomal rearrangements or activate proto-oncogenes, as evidenced by studies showing persistent DNA aberrations even with high-fidelity variants. In editing, absent from approved uses but pursued in , mosaicism and heritable mutations pose amplified risks, with animal models revealing unintended structural variants propagating across generations. Delivery vectors like AAV may trigger immune-mediated clearance or over decades, untested in humans beyond preliminary data. Reviews highlight epistemic gaps, including understudied epigenetic perturbations and interactions with environmental factors, underscoring the need for multi-decade observational studies to quantify risks empirically rather than rely on predictive models. While no causal links to cancer or other late-onset pathologies have been confirmed in edited organisms to date, the absence of does not equate to , given the technology's decade-long timeline.

Ethical and Societal Dimensions

Germline Editing and Heritability Concerns

Germline editing refers to the application of genome editing technologies, such as CRISPR-Cas9, to germ cells, zygotes, or early-stage embryos, resulting in modifications that are incorporated into the heritable genome and transmitted to subsequent generations.00560-3/fulltext) Unlike somatic editing, which affects only the treated individual, germline alterations propagate through the population, amplifying the consequences of any errors or unintended outcomes across familial lineages. A primary concern with heritable editing is the persistence and potential accumulation of off-target effects, where the editing machinery induces unintended at non-target genomic sites. Studies have demonstrated that CRISPR-Cas9 can generate large structural variants, deletions, or insertions both on-target and off-target, with detection challenges in early due to cellular heterogeneity and mosaicism—where not all cells in an carry the same edit. In germline contexts, such could lead to novel genetic disorders in or descendants, as empirical data from model organisms indicate that off-target rates, though reduced by high-fidelity variants, remain non-zero and context-dependent. exacerbates these risks, as a single viable edited implanted for could disseminate alterations unpredictably, without the selective pressures that mitigate harmful in natural . The 2018 case of illustrates these heritability perils empirically. He, a Chinese biophysicist, announced the birth of twin girls whose embryos he edited using to disrupt the gene, aiming to confer HIV resistance by mimicking a natural delta-32 . Subsequent analysis revealed mosaicism in the embryos, incomplete biallelic editing, and potential off-target risks, with no long-term health data available as of 2023; He was convicted in 2019 of illegal medical practice and sentenced to three years in prison. This incident underscored causal uncertainties: while the intended edit might offer partial protection, heritable mosaicism could result in variable expressivity across generations, potentially introducing fitness costs or oncogenic risks not observable in the initial cohort. Regulatory frameworks reflect these concerns, with heritable genome editing prohibited in the vast majority of jurisdictions due to unresolved safety and heritability issues. As of 2020, 75 of 96 surveyed countries explicitly ban the use of genetically modified embryos to initiate , including statutory prohibitions in nations like , , , and the under the Oviedo Convention; no country permits clinical reproduction via edits. International bodies, including a 2019 call from scientific summits, advocate moratoriums until empirical evidence demonstrates negligible off-target rates and predictable intergenerational outcomes, prioritizing causal verification over speculative benefits.01260-0/fulltext) Despite theoretical potential for preventing monogenic diseases, the absence of comprehensive long-term data—coupled with evidence of persistent editing inaccuracies—renders heritable applications empirically unjustified at present.

Enhancement Debates and Equity Issues

The debate over through genome editing distinguishes therapeutic applications, which aim to correct genetic s, from non-therapeutic enhancements that seek to improve traits such as , physical prowess, or resistance beyond normal human variation. Proponents of enhancement, including some bioethicists, contend that individuals should have to pursue genetic improvements, analogous to existing enhancements like or , provided risks are managed and is informed. Critics argue that enhancements risk commodifying human life, eroding genetic diversity, and reviving eugenic practices, with empirical precedents like the 2018 experiment—where embryos were edited for HIV resistance via CCR5 deletion—illustrating blurred lines, as the modification may confer unintended advantages like West Nile virus protection but also potential cognitive drawbacks. These concerns are amplified in germline editing, where changes are heritable, prompting calls for moratoriums from bodies like the until safety and societal impacts are better understood. A central contention is the "" argument, positing that permitting (non-heritable) therapies inevitably leads to enhancements due to and shifting norms, as the boundary between alleviating deficits and optimizing traits proves arbitrary—e.g., for severe versus mild cognitive boosts. Empirical analysis of this slope reveals logical weaknesses, as regulatory distinctions can be maintained through evidence-based criteria, yet historical precedents in reproductive technologies suggest normative drift where initial therapeutic intents expand to elective uses. Philosophically, enhancement challenges egalitarian principles by potentially creating a genetic , with first-principles reasoning highlighting causal risks: unequal adoption could entrench of advantage, as modeled in simulations showing rapid divergence in trait distributions under selective . Despite these debates, no verified human enhancements beyond therapeutic trials exist as of 2025, confining discussions largely to prospective ethics. Equity issues arise from genome editing's high development and delivery costs, restricting access primarily to affluent populations and high-income countries, thereby widening global health disparities. The first approved CRISPR therapy, Casgevy for sickle cell disease and beta-thalassemia, launched in 2023 at $2.2 million per patient, exceeds lifetime conventional treatment costs ($4-6 million for sickle cell) yet remains unaffordable for most, particularly in sub-Saharan Africa where sickle cell prevalence reaches 1-2% of births. This pricing, driven by manufacturing complexities like ex vivo cell editing and chemotherapy conditioning, disadvantages low-resource settings despite the technology's potential to address endemic genetic burdens. For enhancements, equity concerns intensify: if viable, they could exacerbate class divides, as only elites afford "designer" traits, fostering a bifurcated society where genetic haves outcompete have-nots in labor markets and reproduction, per economic models of technological inequality. Proposals for equitable scaling include value-based pricing and international funding mechanisms, but implementation lags, underscoring causal realism that without deliberate access reforms, editing reinforces existing socioeconomic gradients rather than mitigating them.

Regulatory Responses and Innovation Barriers

Following the 2018 revelation of unauthorized germline editing by in , which resulted in the birth of edited infants, international bodies responded with calls for enhanced oversight. The World Health Organization's 2021 governance framework for human genome editing emphasized institutional, national, and global mechanisms to promote safety, transparency, and equity, while advising against heritable edits until scientific, ethical, and societal consensus is achieved. This framework, developed by an expert advisory committee, prioritizes and public engagement but lacks enforceable authority, relying on voluntary adoption by member states. In the United States, the (FDA) oversees genome editing under its human regulations, treating CRISPR-based products as biologics subject to applications and phased clinical trials. A March 2024 FDA guidance specifically addresses human products incorporating genome editing of cells, recommending preclinical assessments for off-target effects, , and to mitigate risks like unintended mutations. editing remains prohibited by congressional acts, with no federal protocols permitting heritable modifications, though applications advanced with the December 2023 approval of Casgevy (exagamglogene autotemcel), the first therapy for and beta-thalassemia. FDA clinical holds, such as the temporary pause on Verve Therapeutics' VERVE-101 in 2022 due to delivery concerns (lifted in October 2023), exemplify precautionary scrutiny that delays trials. The European Union applies its 2001 GMO Directive to genome-edited organisms, classifying most edits as equivalent to transgenic GMOs requiring rigorous risk assessments, labeling, and traceability, which has stifled agricultural innovation by equating precise edits to foreign DNA insertions. A February 2024 European Parliament vote proposed easing regulations for edits without foreign DNA or unintended changes, exempting them from GMO rules to foster competitiveness, but implementation remains pending amid member state opt-outs and environmental NGO opposition. For human applications, heritable editing is banned under the 2001 Clinical Trials Directive and Oviedo Convention protocols ratified by several members. China intensified restrictions post-He Jiankui, issuing a 2019 interim ban on heritable editing and enacting 2021 provisions criminalizing clinical use, followed by a July 2024 Ministry of Science and Technology prohibition on all clinical research as "irresponsible" due to safety uncertainties. These measures, enforced through institutional reviews, contrast with permissive somatic trials but reflect reactive centralization amid prior lax oversight. Such frameworks impose innovation barriers by creating regulatory asymmetry, prompting forum-shopping where researchers and firms relocate to jurisdictions like the or with lighter crop editing rules, reducing European investment in biotech by an estimated 20-30% in affected sectors. Precautionary standards, including lengthy approvals (often 10-15 years for FDA gene therapies) and ambiguity in non-human applications like , deter ; a 2024 analysis linked stringent EU GMO equivalence to fewer commercialized gene-edited products and heightened R&D costs. prohibitions, while addressing heritable risks, foreclose enhancements or population-level interventions, with empirical data showing off-target rates below 1% in optimized systems yet insufficient for policy shifts amid uncertainty. Proponents argue risk-proportionate rules could accelerate benefits, as therapies demonstrate, but institutional inertia—exacerbated by biases toward alarmism in academic and sources—perpetuates barriers over evidence-based .

References

  1. [1]
    Genome-Editing Technologies: Concept, Pros, and Cons of Various ...
    Apr 3, 2019 · Principally, genome-wide editing techniques can be interpreted as methods where DNA sequences are changed by deletions, mRNA processing, and ...
  2. [2]
    Genome Editing: Past, Present, and Future - PMC - NIH
    Dec 19, 2017 · Genome Editing Issues. Remarkably, all that the genome editing nucleases do is to make a break in chromosomal DNA. The key, of course, is that ...
  3. [3]
    History of CRISPR-Cas from Encounter with a Mysterious Repeated ...
    Mar 12, 2018 · To celebrate the 30th anniversary of the discovery of CRISPR, this minireview briefly discusses the fascinating history of CRISPR-Cas systems, ...
  4. [4]
    CRISPR–Cas9: A History of Its Discovery and Ethical ... - NIH
    Aug 15, 2022 · CRISPR-Cas9, or 'genetic scissors', is a genome editing technology that won a Nobel Prize. It protects prokaryotes and is used in medicine and ...
  5. [5]
    CRISPR technology: A decade of genome editing is only ... - Science
    Jan 20, 2023 · This Review covers the origins and successes of CRISPR-based genome editing and discusses the most pressing challenges.
  6. [6]
    THE PROMISE AND CHALLENGE OF THERAPEUTIC GENOME ...
    Genome editing, involving precise manipulation of cellular DNA sequences to alter cell fates and organism traits, offers the potential to both understand ...<|separator|>
  7. [7]
    What are the Ethical Concerns of Genome Editing?
    Aug 3, 2017 · Most ethical discussions about genome editing center on human germline editing because changes are passed down to future generations.
  8. [8]
    Ethical Perspectives of Therapeutic Human Genome Editing ... - NIH
    Nov 27, 2022 · This paper argues that allowing this procedure to be the norm would conflict with the research ethic principle of non-exploitation. Zang and ...
  9. [9]
    Beyond safety: mapping the ethical debate on heritable genome ...
    Apr 20, 2022 · In this article, we explore some of the key categorical as well sociopolitical considerations raised by the potential uses of heritable genome editing ...
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    History of genome editing: From meganucleases to CRISPR
    Feb 23, 2021 · We summarize the history of genome editing platforms, starting from enhanced integration of foreign DNA by meganuclease-mediated double-strand breaks to CRISPR ...<|separator|>
  14. [14]
    The Nobel Prize in Physiology or Medicine 2007 - Advanced ...
    Capecchi, Martin J. Evans and Oliver Smithies for their discoveries of principles for introducing specific gene modifications in mice by the use of embryonic ...
  15. [15]
    Origins of Programmable Nucleases for Genome Engineering - PMC
    We started by engineering two novel chimeric nucleases (which were later renamed as zinc finger nucleases or ZFNs) by fusing two three-finger proteins, Sp1-QNR ...
  16. [16]
    Nucleases in gene-editing technologies: past and prologue
    The foundation of gene editing was established in the 1970s and 1980s when researchers successively performed precise gene modifications in yeast and mammalian ...
  17. [17]
    The discovery of zinc fingers and their development for ... - PubMed
    Zinc fingers were discovered in 1985, arising from the interpretation of our biochemical studies on the interaction of the Xenopus protein transcription factor ...
  18. [18]
    The discovery of zinc fingers and their development for practical ...
    Aug 10, 2025 · Zinc fingers were discovered in 1985, arising from the interpretation of our biochemical studies on the interaction of the Xenopus protein ...
  19. [19]
    Gene knockout and knockin by zinc-finger nucleases
    The invention of zinc-finger nucleases (ZFNs). While the zinc-finger motif was discovered in the 1980s [2], ZFNs have a shorter history. Prior to the invention ...Missing: key | Show results with:key
  20. [20]
    TALE and TALEN genome editing technologies - ScienceDirect
    Back in the 1990s, meganucleases and zinc finger nucleases (ZFNs) laid the groundwork for the concept of genome editing and initiated the development of the ...2. Structure And Function Of... · 3. Genome Editing With Talen · 3.1. Talen Architectures
  21. [21]
    A Tale of How TALENs Launched the Genome Editing Revolution
    Mar 11, 2025 · First identified in 1989 as individual proteins involved in pathogen virulence, TALEs and the response that some plants developed in response to ...
  22. [22]
    TALEN and CRISPR/Cas Genome Editing Systems: Tools of Discovery
    Although TALEN and CRISPR/Cas9 appeared recently, these systems have proved to be effective and reliable tools for genome engineering.
  23. [23]
    A short, idiosyncratic history of genome editing - ScienceDirect
    This article reviews some of the early events leading to the development of the first genome editing platform, zinc-finger nucleases.
  24. [24]
    A Programmable Dual-RNA–Guided DNA Endonuclease ... - Science
    Jun 28, 2012 · Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA- ...
  25. [25]
    Press release: The Nobel Prize in Chemistry 2020 - NobelPrize.org
    Oct 7, 2020 · Since Charpentier and Doudna discovered the CRISPR/Cas9 genetic scissors in 2012 their use has exploded. This tool has contributed to many ...
  26. [26]
    A programmable dual-RNA-guided DNA endonuclease in adaptive ...
    Aug 17, 2012 · Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA- ...
  27. [27]
    CRISPR Timeline | Broad Institute
    Zhang, who had previously worked on other genome editing systems such as TALENs, was first to successfully adapt CRISPR-Cas9 for genome editing in eukaryotic ...
  28. [28]
    CRISPR–Cas9 wins Nobel | Nature Reviews Molecular Cell Biology
    Oct 13, 2020 · That year, Emmanuelle Charpentier and Jennifer Doudna published their landmark paper describing the CRISPR–Cas9 system (Science, 17 Aug 2012).
  29. [29]
    CRISPR Statistics 2025 By Success Rate, Gene Editing, Sequencing
    Similarly, the number of CRISPR-related scientific publications increased from 87 in 2011 to 3,917 in 2018.
  30. [30]
    Broad Institute awarded first patent for engineered CRISPR-Cas9 ...
    Apr 15, 2014 · Zhang is also the senior author of the 2013 Science paper showing that Cas9 can be harnessed to modify DNA in mammalian cells.
  31. [31]
    Past, present, and future of CRISPR genome editing technologies: Cell
    Feb 29, 2024 · Genome editing originally arose from advancements in the field of eukaryotic DNA repair. Pioneering studies in the early 1990s using homing ...
  32. [32]
    The hidden risks of CRISPR/Cas: structural variations and genome ...
    Aug 5, 2025 · CRISPR/Cas technology has revolutionized gene editing by enabling precise and efficient sequence-specific DNA cleavage for targeted genome ...
  33. [33]
    Emerging trends in prime editing for precision genome editing - PMC
    Jul 31, 2025 · Prime editing has rapidly become a versatile tool, supporting a wide range of genetic modifications, including point mutations, insertions and ...
  34. [34]
    Single-Base Editing Seems to Work | Science | AAAS
    Mar 10, 2025 · This was a trial to correct alpha-1 antitrypsin deficiency (AATD), which is a genetically based condition involving a mutation in the SERPINA1 gene.<|separator|>
  35. [35]
    Current advancement in the application of prime editing - Frontiers
    In 2019, David Liu's group developed a new technology named prime editing (PE), which significantly reduced the off-target editing and increased the scope of ...
  36. [36]
    Highly targeted CRISPR delivery advances gene editing in living ...
    Feb 8, 2024 · A new, precision-targeted delivery method for CRISPR-Cas9, published Jan. 11 in the journal Nature Biotechnology, enables gene editing on very specific subsets ...<|separator|>
  37. [37]
    Recent advances in therapeutic gene-editing technologies
    Jun 4, 2025 · This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
  38. [38]
    FDA Approves First Gene Therapies to Treat Patients with Sickle ...
    Dec 8, 2023 · The FDA approved the first cell-based gene therapies, Casgevy and Lyfgenia, for the treatment of sickle cell disease in patients 12 years ...
  39. [39]
    Press Release - CRISPR Therapeutics
    Jan 16, 2024 · The FDA approved CASGEVY, a CRISPR/Cas9 gene-edited cell therapy, for transfusion-dependent beta thalassemia (TDT) in patients 12 and older.
  40. [40]
    Vertex Announces US FDA Approval of CASGEVY ...
    Jan 16, 2024 · Vertex Announces US FDA Approval of CASGEVY™ (exagamglogene autotemcel) for the Treatment of Transfusion-Dependent Beta Thalassemia · severe ...
  41. [41]
    CRISPR Therapeutics Highlights Strategic Priorities and Anticipated ...
    Jan 13, 2025 · 2025 is an important inflection year for CRISPR Therapeutics as we establish CASGEVY and advance our pipeline candidates to human proof-of-concept and beyond.Missing: 2021-2025 | Show results with:2021-2025
  42. [42]
    CRISPR-based Gene Editing Market | Industry Report, 2033
    The global CRISPR-based gene editing market size was estimated at USD 4.01 billion in 2024 and is projected to reach USD 13.50 billion by 2033, ...<|separator|>
  43. [43]
    CRISPR Clinical Trials: A 2025 Update - Innovative Genomics Institute
    Jul 9, 2025 · An update on the progress of CRISPR clinical trials with the latest data and a survey of the CRISPR landscape in 2025.
  44. [44]
    DNA repair pathway choices in CRISPR-Cas9 mediated genome ...
    The two major pathways are error-free, template-dependent homologous recombination (HR), and the error-prone, template-independent classic non-homologous end ...
  45. [45]
    Harnessing DSB repair to promote efficient homology-dependent and
    Mar 24, 2022 · Here we exploit DNA double-strand break (DSB) repair to develop two strategies that install precise genomic insertions using an SpCas9 nuclease-based prime ...
  46. [46]
    Simultaneous inhibition of DNA-PK and Polϴ improves integration ...
    Aug 14, 2023 · Our study reveals DNA-dependent protein kinase (DNA-PK) as the most effective target to improve CRISPR/Cas9-mediated insertions, confirming previous findings.
  47. [47]
    Microhomology-mediated end-joining-dependent integration of ...
    Nov 20, 2014 · MMEJ is a DSB repair mechanism that uses microhomologous sequences (5–25 bp) for error-prone end-joining. In the cell cycle, MMEJ repair is ...
  48. [48]
    Modulation of the microhomology-mediated end joining pathway ...
    Apr 29, 2024 · We found the microhomology-mediated end joining (MMEJ) DNA repair pathway plays a predominant role in Cas9-induced large deletions (LDs).
  49. [49]
    Microhomology-mediated end joining: a back-up survival ...
    Microhomology-mediated end joining (MMEJ) is an error-prone repair mechanism that involves alignment of microhomologous sequences internal to the broken ends ...
  50. [50]
    Genome-Editing Technologies: Principles and Applications - PMC
    Targeted nucleases have provided researchers with the ability to manipulate virtually any genomic sequence, enabling the facile creation of isogenic cell ...
  51. [51]
    Precise genome-editing in human diseases: mechanisms, strategies ...
    Feb 26, 2024 · Precise genome-editing platforms are versatile tools for generating specific, site-directed DNA insertions, deletions, and substitutions.
  52. [52]
    Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR ...
    Each zinc finger domain recognizes a 3- to 4-bp DNA sequence, and tandem domains can potentially bind to an extended nucleotide sequence (typically with a ...
  53. [53]
    Meganucleases and Other Tools for Targeted Genome Engineering
    Nuclease-based approaches, wherein an endonuclease is used to trigger site-specific genome editing, can significantly increase the percentage of targeted cells.
  54. [54]
    Delivery methods for site-specific nucleases: Achieving the full ...
    Meganucleases, or homing endonucleases, are naturally occurring endonucleases that recognize 12 to 40 nucleotide sequences with high specificity [46]. This ...
  55. [55]
    Salient Features of Endonuclease Platforms for Therapeutic ...
    Feb 16, 2016 · Meganucleases are unique among the gene-editing platforms in that their DNA binding and cleavage activities are integrated. The DNA target can ...
  56. [56]
    the Future: Pre-CRISPR Systems Are Driving Therapies to the Clinic
    Feb 7, 2019 · TALENs and ZFNs—the genome editing systems that preceded CRISPR—are looking new again as clinical work shows off their unique advantages.<|separator|>
  57. [57]
    Engineering of customized meganucleases via in vitro ...
    In this chapter, we describe an approach to retarget meganucleases to DNA targets of interest (such as sequences found in genes and cis regulatory regions)
  58. [58]
    Redesign of extensive protein–DNA interfaces of meganucleases ...
    Specific gene-targeting nucleases are critical for genetic engineering and therapy. Meganucleases have been shown to harbor many properties, including small ...
  59. [59]
    Efficient design of meganucleases using a machine learning approach
    Jun 17, 2014 · Meganucleases are important tools for genome engineering, providing an efficient way to generate DNA double-strand breaks at specific loci ...<|separator|>
  60. [60]
    Advancements in genome editing tools for genetic studies and crop ...
    Feb 2, 2025 · Meganucleases require the ability to design nucleases with a high level of sequence specificity to achieve exact genome modification. However, ...
  61. [61]
    Genome editing with engineered zinc finger nucleases - Nature
    Here we review the development of site-specific ZFNs and the application of these reagents to genome editing. We describe methods for engineering ZFNs and ...
  62. [62]
    Genome Engineering With Zinc-Finger Nucleases - PMC - NIH
    Zinc-finger nucleases (ZFNs) are targetable DNA cleavage reagents that have been adopted as gene-targeting tools. ZFN-induced double-strand breaks are ...Missing: milestones | Show results with:milestones
  63. [63]
    Diversifying the structure of zinc finger nucleases for high-precision ...
    Mar 8, 2019 · Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. 19, 1279–1288 (2009) ...
  64. [64]
    Recent developments and clinical studies utilizing engineered zinc ...
    Zinc finger nucleases (ZFNs) are custom-designed artificial endonucleases that make DSBs at specific sequences and have been used as a tool for targeted genetic ...
  65. [65]
    Sangamo Receives Fast Track Designation From The FDA For SB ...
    Jul 13, 2017 · The US Food and Drug Administration (FDA) has granted Fast Track designation to SB-318 and SB-913, the Company's clinical stage in vivo genome editing product ...
  66. [66]
    First-in-human in vivo genome editing via AAV-zinc-finger nucleases ...
    Oct 25, 2022 · ZFN in vivo editing therapy was well tolerated at doses up to 5e13 vg/kg, the highest dose tested. Thirty-three treatment-related adverse events ...
  67. [67]
    Zinc finger nuclease-mediated gene editing in hematopoietic stem ...
    Oct 16, 2024 · In these studies, we show that ZFN-mediated editing is highly efficient and produces HSC capable of long-term multilineage engraftment in vivo, ...
  68. [68]
    Current landscape of gene‐editing technology in biomedicine ...
    Jul 14, 2022 · Many advantages, including low immunogenicity and appropriate gene size, have prompted the widespread use of ZFNs as gene-editing tools for ...<|separator|>
  69. [69]
    Recent advances in the use of ZFN-mediated gene editing for ... - NIH
    This potentially has two drawbacks: 1) the plasmids continue to express the ZFNs that accumulate in high levels in cells, promoting promiscuous DNA binding and ...Missing: disadvantages | Show results with:disadvantages
  70. [70]
    Pros and cons of ZNFs, TALENs, and CRISPR/Cas
    ZNFs use DNA triplets, TALENs use single nucleotides, and CRISPR/Cas uses RNA. CRISPR/Cas is efficient, but all have off-site effects and mosaicism.
  71. [71]
    TALENs—an indispensable tool in the era of CRISPR: a mini review
    Aug 21, 2021 · TALEs proteins were first reported in 2009, derived from phytopathogenic bacterial genus Xanthomonas [26]. TALE is a special class of proteins ...
  72. [72]
    [PDF] Transcription Activator Like Effector Nucleases (TALENs)
    This database gives you the option of selecting a range of dates (TALENs were first reported in 2010) and the ability to select entries from the relevant ...
  73. [73]
    TALENs | Summary - WhatisBiotechnology.org
    TALENs: timeline of key events ; 5 Nov 2015, First successful use of gene therapy to treat baby dying from leukaemia, Vehs, Quasim ; 15 Apr 2016, Gene editing ...Missing: history | Show results with:history
  74. [74]
    A TALEN genome editing system to generate human stem cell ... - NIH
    TALENs are engineered nucleases used to generate mutant alleles of genes in human stem cells, creating disease models. They are assembled in 1-2 days.Missing: history | Show results with:history
  75. [75]
    The comparison of ZFNs, TALENs, and SpCas9 by GUIDE-seq in ...
    Compared to ZFNs, the specificity of TALENs was considered better than that of ZFNs. Still, different TALEN target sites exhibited distinct off-target profiles.
  76. [76]
    [PDF] Expanding the genetic editing tool kit: ZFNs, TALENs, and CRISPR ...
    Zinc finger nucleases (ZFNs) are increasingly being used in academic and industry research for a variety of purposes ranging from the generation of animal ...<|separator|>
  77. [77]
    TALENs: a widely applicable technology for targeted genome editing
    To date, TALENs have been used to make knockout mutations in Arabidopsis and to introduce resistance to infection by Xanthomonas bacteria in rice by disrupting ...
  78. [78]
    TALENs—an indispensable tool in the era of CRISPR: a mini review
    TALEN, on the other hand, has the ability to efficiently manipulate mtDNA (mitochondrial DNA) as a treatment for treating human mitochondrial diseases triggered ...
  79. [79]
    CRISPR-cas system: biological function in microbes and its use to ...
    Jul 5, 2019 · The CRISPR-cas system is an adaptive immune system of bacteria and archaea, which protects the bacteria from invaders, including bacteriophages ...
  80. [80]
    CRISPR-Cas: biology, mechanisms and relevance - PMC - NIH
    Type II CRISPR-Cas systems seem to play a significant role in regulating virulence of pathogenic bacteria. In Francisella novicida, a ribonucleoprotein complex ...
  81. [81]
    CRISPR in Nature - Innovative Genomics Institute (IGI)
    Sep 12, 2022 · CRISPR systems evolved to defend bacteria against bacterial viruses called phages. Learn how phages infect bacteria and how those bacteria use CRISPR and other ...
  82. [82]
    Origins and evolution of CRISPR-Cas systems - Journals
    Mar 25, 2019 · The CRISPR-Cas systems consist of distinct adaptation and effector modules whose evolutionary trajectories appear to be at least partially independent.
  83. [83]
    What is CRISPR: Your Ultimate Guide | Synthego
    The CRISPR system is the basis of adaptive immunity in bacteria and archaea. It utilizes Cas nucleases, which are enzymes that can bind and create double ...Crispr Methods And... · Crispr In Disease Models And... · 7 Prominent Crispr...
  84. [84]
    Multiplex Genome Engineering Using CRISPR/Cas Systems - Science
    We engineered two different type II CRISPR/Cas systems and demonstrate that Cas9 nucleases can be directed by short RNAs to induce precise cleavage at ...
  85. [85]
    Multiplex Genome Engineering Using CRISPR/Cas Systems - PMC
    Functional elucidation of causal genetic variants and elements requires precise genome editing technologies. The type II prokaryotic CRISPR (clustered ...
  86. [86]
    CRISPR Guide - Addgene
    The CRISPR-Cas complex recruits the transposase proteins to the target location in the genome, where the transposase then carries out integration of donor DNA.
  87. [87]
    Importance of the PAM Sequence in CRISPR Experiments - Synthego
    The PAM is required for a Cas nuclease to cut and is generally found 3-4 nucleotides downstream from the cut site.
  88. [88]
    A Review on the Mechanism and Applications of CRISPR/Cas9 ...
    Sep 27, 2022 · This review aims to provide detail on two classes (class 1 and 2) of the CRISPR/Cas system, and the mechanisms of all Cas proteins, including Cas12, Cas13, and ...
  89. [89]
    CRISPR 101: Cas9 vs. The Other Cas(s) - Addgene Blog
    Dec 8, 2022 · Cas13 is an RNA-guided nuclease, however, unlike Cas9, it targets RNA instead of DNA. This makes it particularly useful in generating transient ...<|control11|><|separator|>
  90. [90]
    CRISPR-Cas: Converting A Bacterial Defence Mechanism into A ...
    This review illustrates the mechanisms behind CRISPR-Cas systems as a means of bacterial immunity against phage invasion and how these systems were engineered ...
  91. [91]
    Assessing and advancing the safety of CRISPR-Cas tools: from DNA ...
    Jan 13, 2023 · Among them, most Cas9 and Cas12 variants possess DNA endonuclease activity, while Cas13 variants show preferential RNA-targeting and cleavage ...
  92. [92]
    Base editing: precision chemistry on the genome and transcriptome ...
    Oct 15, 2018 · Base editing is a newer genome-editing approach that uses components from CRISPR systems together with other enzymes to directly install point mutations into ...
  93. [93]
    Programmable editing of a target base in genomic DNA without ...
    A new approach to genome editing that enables the direct, irreversible conversion of one target DNA base into another in a programmable manner.
  94. [94]
    Base editing: advances and therapeutic opportunities - Nature
    Oct 19, 2020 · In this study, BE-PLUS demonstrated an average 5.6-fold decrease in indel formation and 3.6-fold decrease in C•G to non-T•A editing across seven ...
  95. [95]
    Programmable base editing of A•T to G•C in genomic DNA without ...
    Nov 1, 2017 · Now, David Liu and collaborators describe the next step in genomic base editing technology, designed to tackle the conversion of A•T base ...
  96. [96]
    From bench to bedside: cutting-edge applications of base editing ...
    Dec 20, 2024 · This article reviews the current progress of base editors and prime editors, elaborating on specific examples of their applications in the therapeutic field.
  97. [97]
    Search-and-replace genome editing without double-strand ... - Nature
    Oct 21, 2019 · Here we describe prime editing, a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site.
  98. [98]
    Prime editing for precise and highly versatile genome manipulation
    In this Review, Chen and Liu discuss the latest developments in prime editing systems, including improvements to their editing efficiency and capabilities, as ...<|separator|>
  99. [99]
    ProPE expands the prime editing window and enhances gene ...
    Oct 10, 2025 · Prime editing (PE), which requires neither the generation of DNA double-strand breaks nor the use of donor DNA, reduces the production of ...
  100. [100]
    David Liu Wins 2025 Breakthrough Prize for Base Editing and Prime ...
    Apr 5, 2025 · David Liu, the inventor of base editing and prime editing, has been awarded the 2025 Breakthrough Prize in Life Sciences.
  101. [101]
    A laboratory-evolved CRISPR-associated transposase adapts to ...
    Jun 16, 2025 · CRISPR-associated transposases (CASTs) are an attractive candidate for genome editing applications, as they enable the insertion of large DNA cargoes without ...
  102. [102]
    Drag-and-drop genome insertion of large sequences without double ...
    Nov 24, 2022 · We present programmable addition via site-specific targeting elements (PASTE), which uses a CRISPR–Cas9 nickase fused to both a reverse ...
  103. [103]
    Unlocking the potential of CRISPR-associated transposons - PubMed
    May 19, 2025 · CRISPR-associated transposons (CASTs) are emerging genome-editing tools that enable RNA-guided DNA integration without inducing double-strand breaks (DSBs).
  104. [104]
  105. [105]
    Bacterial Retrons Enable Precise Gene Editing in Human Cells - PMC
    Retrons, which reverse transcribe RNA into msDNA, are used with CRISPR-Cas9 to produce msDNA as donor DNA for precise gene editing in human cells.
  106. [106]
  107. [107]
    Programming cells by multiplex genome engineering and ... - NIH
    MAGE provides a highly efficient, inexpensive and automated solution to simultaneously modify many genomic locations (for example, genes, regulatory regions) ...
  108. [108]
    Multiplex genome engineering in eukaryotes - Nature
    Nov 27, 2017 · Multiplex automated genome engineering (MAGE) has been used in prokaryotes to simultaneously target many locations on the chromosome for ...
  109. [109]
    Viral and Non-Viral Systems to Deliver Gene Therapeutics to ... - NIH
    Jul 4, 2024 · In this review, we describe the different delivery systems available for genome editing components, including viral and non-viral systems, highlighting their ...
  110. [110]
    Viral vector platforms within the gene therapy landscape - Nature
    Feb 8, 2021 · In this review, we will describe three viral vector platforms that have gained wide use for efficacious gene therapy and regulatory approval.
  111. [111]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · Since 2013, explosive growth has been recorded in the study of CRISPR technology, with tens of thousands of CRISPR-related articles published.
  112. [112]
    Advanced delivery systems for gene editing - ScienceDirect.com
    We described the plethora of delivery systems available for genome editing components, including viral and non-viral systems, highlighting their advantages, ...Review · Introduction · Delivery Of Gene-Editing...
  113. [113]
    Customizable virus-like particles deliver CRISPR–Cas9 ... - Nature
    Feb 10, 2025 · Here we develop a cell-tropism programmable CRISPR–Cas9 ribonucleoprotein delivery system (RIDE) based on virus-like particles.
  114. [114]
    Applications and challenges of CRISPR-Cas gene-editing to ...
    The ex vivo gene editing method has three main potential applications in clinics: cancer immunotherapy, treatment of hereditary diseases (e.g. sickle cell ...
  115. [115]
    Genome Editing Therapy for the Blood: Ex Vivo Success and In Vivo ...
    Here we review recent advances in therapeutic genome editing of HSCs and T cells, focusing on the progress in ex vivo contexts, the promise of improved access ...
  116. [116]
    Advantages and challenges of ex vivo generation and ... - PubMed
    Mar 12, 2025 · Despite advancements in this field, two primary challenges persist: low differentiation efficiency and difficulties in isolating and enriching ...
  117. [117]
    Editorial: Ex-vivo and in-vivo genome engineering for metabolic and ...
    Jul 5, 2023 · Discuss the progress and challenges of ex vivo genome editing in HSPCs to treat non-hematological disorders. One of these challenges is ...
  118. [118]
    CRISPR Clinical Trials: A 2024 Update - Innovative Genomics Institute
    Mar 13, 2024 · In late 2023, we saw the first-ever approval of CRISPR-based medicine: Casgevy, a cure for sickle cell disease (SCD) and transfusion-dependent beta thalassemia ...Missing: patents | Show results with:patents
  119. [119]
    Landscape of ex vivo gene therapies: Technological trends and ...
    In vivo gene therapy delivers new or corrected genes using a vector directly into the body, either systemically or locally. The second approach, ex vivo gene ...
  120. [120]
    Therapeutic in vivo delivery of gene editing agents - ScienceDirect
    Jul 21, 2022 · In vivo gene editing, where cells are edited directly within the body, offers the greatest promise for treating genetic disorders. However, in ...
  121. [121]
    Revolutionizing in vivo therapy with CRISPR/Cas genome editing
    Researchers are investigating the ability of CRISPR/Cas9 to precisely edit genes and address specific mutations and the potential to provide new cures and ...Missing: post- | Show results with:post-
  122. [122]
    CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis
    Jun 26, 2021 · We report evidence of CRISPR-Cas9–based in vivo gene editing in humans. Systemic administration of NTLA-2001 to six patients with hATTR ...Missing: examples | Show results with:examples
  123. [123]
    How Scientists Used CRISPR to Edit Human DNA In Vivo
    Apr 8, 2020 · This video explains the principles of DNA editing with CRISPR-Cas9 and the rationale for its use and testing as a therapy for LCA10 in the BRILLIANCE clinical ...Missing: examples | Show results with:examples
  124. [124]
    In vivo delivery of CRISPR-Cas9 genome editing components for ...
    The first clinical trial of CRISPR therapeutics in humans was initiated in 2016, aiming to use CRISPR-Cas9 engineered PD-1 knockout T cells for the treatment of ...
  125. [125]
    The comparison of ZFNs, TALENs, and SpCas9 by GUIDE-seq in ...
    We compared the efficiency and specificity of ZFNs, TALENs, and SpCas9 by GUIDE-seq. SpCas9 outperformed ZFNs and TALENs with higher efficiency and specificity.
  126. [126]
    Off-target Effects in CRISPR/Cas9-mediated Genome Engineering
    Here, we review the basic mechanisms underlying off-target cutting in the CRISPR/Cas9 system, methods for detecting off-target mutations, and strategies for ...
  127. [127]
    Minimizing the off-target frequency of the CRISPR/Cas9 system via ...
    Jun 14, 2023 · The rates of mutation at the selected off-target sites were very high, ranging from 9.4% to 93.6% when the cells were edited using native CRISPR ...
  128. [128]
    Off-target effects in CRISPR/Cas9 gene editing - PMC - NIH
    A major concern in the applications of the CRISPR/Cas9 system is about its off-target effects, namely the deposition of unexpected, unwanted, or even adverse ...
  129. [129]
    Recent Advancements in Reducing the Off-Target Effect of CRISPR ...
    Jan 18, 2024 · In this review, we summarize the recent technological advancements in reducing the off-target effect of CRISPR-Cas9 genome editing.
  130. [130]
    Beyond the promise: evaluating and mitigating off-target effects in ...
    Each method has its own set of advantages and limitations; hence, no single method can provide a comprehensive assessment of CRISPR-Cas associated off-targets.Abstract · Introduction · Assessment of CRISPR... · Approaches to reduce off...
  131. [131]
    Improvement of base editors and prime editors advances precision ...
    Base and prime editors are genome engineering tools that introduce precise edits without DSBs, advancing plant research and crop breeding.
  132. [132]
    Emerging trends in prime editing for precision genome editing - Nature
    Jul 31, 2025 · Prime editing is a novel 'search-and-replace' genome editing technology that significantly advances the capabilities of the CRISPR system by ...
  133. [133]
    High-fidelity CRISPR-Cas9 variants with undetectable genome-wide ...
    Here we describe SpCas9-HF1, a high-fidelity variant harboring alterations designed to reduce non-specific DNA contacts.
  134. [134]
    Sniper2L is a high-fidelity Cas9 variant with high activity - Nature
    Mar 9, 2023 · We developed an improved version of Sniper–Cas9, Sniper2L, which represents an exception to this trade-off trend as it showed higher specificity with retained ...
  135. [135]
    Methods for Optimizing CRISPR-Cas9 Genome Editing Specificity
    Aug 4, 2016 · Selecting the most unique target site possible is thus a valuable strategy for improving specificity, because many gene editing applications ...<|separator|>
  136. [136]
    Single-Base Resolution: Increasing the Specificity of the CRISPR ...
    Mar 3, 2021 · Alternative Methods for Specificity Enhancement. Restriction of Cas nuclease abundance is a rational approach to increase the specificity of ...
  137. [137]
    Methodologies for Improving HDR Efficiency - Frontiers
    In order to modify the gene precisely, numerous methods arose by inhibiting NHEJ or enhancing HDR, such as chemical modulation, synchronized expression, and ...Overview of CRISPR-Cas9... · HDR Pathway · Favoring the HDR Pathway...
  138. [138]
    3 Tips to Improve HDR Efficiency for CRISPR Editing in Human Cells
    Sep 5, 2017 · 1. Increasing HDR accuracy with CRISPR/Cas-blocking mutations · 2. Optimizing "cut-to-mutation distance" · 3. Optimizing "cut-to-mutation distance ...
  139. [139]
    Enrichment strategies to enhance genome editing
    Jul 1, 2023 · Reducing the size of the editing window with newer base editors to increase specificity might in turn reduce editing efficiency and limit the ...
  140. [140]
    Enhancing homology-directed repair efficiency with HDR-boosting ...
    Aug 10, 2024 · By combining with an inhibitor of NHEJ or the HDRobust strategy, the modular ssDNA donors achieved 66.62%–90.03% HDR efficiency, offering a ...
  141. [141]
    CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β ...
    Dec 7, 2020 · CRISPR-Cas9 Gene Editing for SCD and TDT. Transfusion-dependent β-thalassemia (TDT) and sickle cell disease (SCD) are the most common monogenic ...
  142. [142]
    Clinical applications of the CRISPR/Cas9 genome-editing system
    CRISPR/Cas9 is used to treat cancers, hemolytic diseases, immunodeficiency, cardiovascular, visual, neurodegenerative, and some X-linked disorders.
  143. [143]
    Overview CRISPR Clinical Trials 2025 - Learn | Innovate | Access
    Feb 28, 2025 · In 2023, therapeutic gene editing moved from promise to reality with the first approved CRISPR-based therapy, CASGEVY, which has since received ...Missing: ex | Show results with:ex
  144. [144]
    Advancing CRISPR genome editing into gene therapy clinical trials
    Ongoing research explores the potential of CRISPR technology for cancer therapies, HIV treatment and other complex diseases.
  145. [145]
    FDA Approves Vertex & CRISPR's Gene Editing Therapy | CAF
    The FDA has approved Vertex and CRISPR's gene-editing therapy Casgevy to treat Beta Thalassemia. Casgevy becomes the first therapy for the rare blood ...
  146. [146]
    Recent advances of CRISPR-based genome editing for enhancing ...
    Sep 22, 2024 · This review highlights the transformative potential of CRISPR/Cas technology, emphasizing recent innovations such as prime and base editing, and ...
  147. [147]
    Adoption of CRISPR-Cas for crop production: present status and ...
    Jun 7, 2024 · So far, more than 25 plant species and 100 genes have been successfully edited using CRISPR-Cas9 to achieve a range of desirable traits in ...
  148. [148]
    Application of CRISPR/Cas-based gene-editing for developing ...
    Aug 16, 2024 · ... yielding better banana with enhanced nutritional content and disease resistance. By integrating precision genetics with traditional breeding ...
  149. [149]
    Engineering crops of the future: CRISPR approaches to develop ...
    Nov 30, 2020 · Here, we discuss the latest developments in CRISPR technologies for engineering resistance to viruses, bacteria, fungi, and pests.<|separator|>
  150. [150]
    CRISPR-Cas9 genome editing in crop breeding for climate change ...
    Recently, a CRISPR-Cas9 system was utilized to edit gibberellin (GA) phytohormone-related mutants that confer a shorter stature to the barley plant [87].
  151. [151]
    A Critical Review: Recent Advancements in the Use of CRISPR ...
    The CRISPR/Cas9 has been used to improve the monocots and dicots crops for a variety of traits like yield, quality, disease resistance, and resistance to ...
  152. [152]
    CRISPR-Cas9 Application in Crop Biotechnology: A Decade of ...
    Aug 13, 2025 · Studies that focused on CRISPR-Cas9 edited crops showed improvement of β-carotene with its sixfold increase in rice and banana, GABA content ...
  153. [153]
    Gene-Edited Plants: Regulation and Issues for Congress
    Sep 7, 2023 · The rule revised APHIS's regulations governing certain gene-edited plants (7 C.F.R. §340) and new genetically engineered plants. It exempts ...
  154. [154]
    [PDF] Plants produced using new genomic techniques
    Oct 3, 2023 · On 5 July 2023, the Commission tabled a proposal for a regulation on certain new genomic techniques (NGTs). It establishes two categories of ...
  155. [155]
    CRISPR in Agriculture: 2024 in Review - Innovative Genomics Institute
    Dec 10, 2024 · CRISPR is being used in a wide breadth of ways in plant and animal agriculture, from reducing waste to adapting plants and animals to climate change.
  156. [156]
    Revolutionizing Agriculture With CRISPR Technology: Applications ...
    Sep 11, 2025 · In summary, CRISPR enables precise genetic improvements in crops, spanning stress tolerance, yield, nutrition, and disease resistance.
  157. [157]
    CRISPR in Animals and Animal Models - ScienceDirect.com
    CRISPR-Cas9 has revolutionized animal genome editing, enabling genetic modification of embryonic stem cells and zygotes, and is used for gene inactivation and ...
  158. [158]
    Generation of gene-edited rats by delivery of CRISPR/Cas9 protein ...
    Nov 29, 2017 · We report the optimization of an electroporation method for intact rat zygotes using sgRNAs and Cas9 protein in combination or not with ssODNs (~100 nt).
  159. [159]
    CRISPR/Cas9: a powerful genetic engineering tool for establishing ...
    Aug 4, 2015 · Here we discuss the use of CRISPR/Cas9 technology to establish large animal models that can more faithfully mimic human neurodegenerative diseases.
  160. [160]
    Redefining mouse transgenesis with CRISPR/Cas9 genome editing ...
    Feb 28, 2018 · A new study shows that the in situ delivery of CRISPR/Cas9 reagents into pregnant mice results in a high efficiency of editing, and enables the rapid ...
  161. [161]
    The Progress of CRISPR/Cas9-Mediated Gene Editing in ...
    Here we review both mice and zebrafish models of human skeletal diseases generated by CRISPR/Cas9 system, and their contributions to deciphering the underpins ...
  162. [162]
    Base editors in zebrafish: a new era for functional genomics ... - NIH
    May 21, 2025 · Base editing has revolutionized genome engineering by enabling precise single-nucleotide modifications without inducing double-strand breaks ...
  163. [163]
    CRISPR-Cas9 in the Tailoring of Genetically Engineered Animals
    CRISPR-Cas9 is used for targeted genome editing in animals, creating models for disease, protein production, and xenotransplantation, and improving livestock ...
  164. [164]
    Use of CRISPR/Cas9 to model brain diseases - ScienceDirect
    In this review, we focus on genomic editing tools that enable the generation of animal models of brain diseases by modifying endogenous genomes.<|separator|>
  165. [165]
    CRISPR-based functional genomics tools in vertebrate models
    Jul 31, 2025 · In popular vertebrate models, including mice and zebrafish, CRISPR has been adapted to high-throughput mutagenesis workflows, knock-in alleles ...
  166. [166]
    Microbial Genome Editing with CRISPR–Cas9: Recent Advances ...
    Overall, combined CRISPR–Cas9 platforms streamline the engineering of these species into microbial factories for amino acids, enzymes, solvents, and biofuels.
  167. [167]
    CRISPR-based gene editing technology and its application in ...
    We summarize recent developments in the application of CRISPR-based gene editing tools for industrial microorganism and probiotics modifications.
  168. [168]
    CRISPR-Made Greener Products and Processes in Industrial ...
    Apr 3, 2023 · One of the major applications of CRISPR in industrial biotechnology is the engineering of enzymes. CRISPR-guided DNA polymerases can be used to ...
  169. [169]
    Vertex and CRISPR Therapeutics Announce US FDA Approval of ...
    Dec 8, 2023 · “CASGEVY's approval by the FDA is momentous: it is the first CRISPR-based gene-editing therapy to be approved in the U.S.
  170. [170]
    Revolutionary breakthrough: FDA approves CASGEVY, the first ...
    Casgevy is the first FDA-approved CRISPR/Cas9 gene therapy for sickle cell disease, offering a curative approach and eliminating the need for transfusions.<|separator|>
  171. [171]
    The first human trial of CRISPR-based cell therapy clears safety ...
    Aug 25, 2020 · Lu et al. examined the feasibility and safety of using CRISPR-engineered T cells to treat late-stage lung cancer. They demonstrated very low off-target editing ...
  172. [172]
    First Chinese CRISPR gene therapy trial demonstrates safety
    Apr 28, 2020 · CRISPR gene therapy appears to be feasible and safe, and the stage is set for the next significant advances in cancer treatment.
  173. [173]
    First U.S. Patients Treated With CRISPR As Human Gene-Editing ...
    Apr 16, 2019 · There's now a clinical trial underway at the University of Pennsylvania using CRISPR for cancer treatment. It involves removing immune system ...
  174. [174]
    First-in-human Phase 1 CRISPR Gene Editing Cancer Trials - PubMed
    A prospective first-in-human Phase 1 CRISPR gene editing trial in the United States for patients with melanoma, synovial sarcoma, and multiple myeloma
  175. [175]
    OHSU performs first-ever CRISPR gene editing within human body
    Mar 4, 2020 · The gene-editing tool CRISPR has been used to address a blindness-causing gene mutation at Oregon Health & Science University for a clinical trial.
  176. [176]
    Landmark CRISPR trial shows promise against deadly disease
    Jun 29, 2021 · Preliminary results from a landmark clinical trial suggest that CRISPR–Cas9 gene editing can be deployed directly into the body to treat disease.
  177. [177]
    CASGEVY™ (gene editing) – TIF
    FDA has accepted Priority Review for BLA submission and set a PDUFA (the date by which the FDA must respond to the application) target date of 8 December 2023.
  178. [178]
    Pipeline | CRISPR Therapeutics
    Exa-cel, a CRISPR/Cas9 gene-edited therapy arising out of our collaboration with Vertex Pharmaceuticals Incorporated, has now been approved in some countries ...
  179. [179]
    World's First Patient Treated with Personalized CRISPR Gene ...
    May 15, 2025 · A child diagnosed with a rare genetic disorder has been successfully treated with a customized CRISPR gene editing therapy by a team at Children's Hospital of ...
  180. [180]
    CRISPR Therapeutics to Present Late-Breaking Data at the ...
    Sep 9, 2025 · Abstract Title: First-in-Human Phase 1 Clinical Trial of a CRISPR-Cas9 Gene Editing Therapy Targeting ANGPTL3 ; Abstract Number: 4392851 ; Session ...
  181. [181]
    Press Release - CRISPR Therapeutics
    The Company plans to present the CTX310 Phase 1 data at a medical meeting in the second half of 2025. CTX320 is in an ongoing Phase 1 clinical trial targeting ...<|separator|>
  182. [182]
    Research and Pipeline | Editas Medicine
    Editas Medicine works with two distinct CRISPR nucleases: Cas9 and Cas12a (also known as Cpf1). Both nucleases have distinct gene editing and targeting ...
  183. [183]
    Quarterly Industry Landscape Report - ASGCT
    The data show unequivocally that gene, cell, and RNA therapies continue to grow rapidly with nearly 3,500 therapies in preclinical and clinical development ...
  184. [184]
    CRISPR Clinical Trials to Follow - Synthego
    Apr 1, 2025 · CRISPR therapies are being developed for genetic disorders, cancer, and infectious diseases using various CRISPR nucleases like eSpOT-ON, ...
  185. [185]
    Exploring the Potential and Challenges of CRISPR Delivery and ...
    May 27, 2024 · The review outlines CRISPR's historical progression and highlights advancements in gene editing methods and delivery vehicles.
  186. [186]
    CRISPR Cell and Gene Therapies: Key Challenges in Clinical ...
    Feb 28, 2025 · Researchers face many hurdles when developing CRISPR therapy products, including the procurement of true GMP reagents, unclear regulatory ...
  187. [187]
    Regulatory Trends in Cell and Gene Therapies - Synthego
    Jun 28, 2022 · Researchers face many hurdles when developing CRISPR therapy products, including the procurement of true GMP reagents, unclear regulatory ...Recent Problems In Cell &... · Expected Growth In Cell And... · Regulatory Harmonization And...
  188. [188]
    Moving Therapeutic Genome Editing into Global Clinical Trials and ...
    May 22, 2025 · Moving CRISPR-based therapies from discovery to dosing patients in clinical trials and ultimately to approval involves navigating a challenging ...
  189. [189]
    Non-clinical safety considerations on genome editing using the ...
    Jul 28, 2025 · Genome editing technologies are rapidly being adopted in clinical trials; however, critical non-clinical safety considerations are required to ...Review Article · Advances In Crispr/cas Gene... · Crispr/cas9-Associated...
  190. [190]
    Key challenges in bringing CRISPR-mediated somatic cell therapy ...
    Sep 25, 2017 · We identify some of the major challenges for clinicians, regulators, and human research ethics committees in the clinical translation of CRISPR-mediated ...
  191. [191]
    Past, present, and future of CRISPR genome editing technologies
    Feb 29, 2024 · In this review, we discuss the current state of CRISPR gene editing technologies in both research and therapy, highlighting limitations that constrain them.
  192. [192]
    Mosaicism in CRISPR/Cas9-mediated genome editing - ScienceDirect
    Jan 15, 2019 · CRISPR/Cas9-mediated mosaic mutations can result following DNA breaks and indel formations (via NHEJ) or oligonucleotide or transgene insertions ...
  193. [193]
    Cas9 immunity creates challenges for CRISPR gene editing therapies
    Aug 29, 2018 · Preventing the immune destruction of CRISPR-Cas9-corrected cells could be more challenging in patients that already have pre-existing anti-Cas9 ...
  194. [194]
    Efficacy and long-term safety of CRISPR/Cas9 genome editing in the ...
    Mar 25, 2021 · One major long-term safety concern is tumorigenesis that might arise if off-targets involve oncogenes and tumor suppressor genes. It is known ...
  195. [195]
    CRISPR-Cas9: The gene editing tool changing the world
    Jan 15, 2025 · While Casgevy has certainly demonstrated CRISPR can be safe and effective as a treatment, it will definitely take some time and additional ...<|separator|>
  196. [196]
  197. [197]
    Societal and Ethical Impacts of Germline Genome Editing: How Can ...
    Oct 9, 2019 · Genome editing has opened up the possibility of heritable alteration of the human germline. The potential of this powerful tool has spurred ...
  198. [198]
    CRISPR-Cas9 induces large structural variants at on-target and off ...
    Feb 2, 2022 · Such off-target mutations can have serious consequences as they might disrupt the function or regulation of non-targeted genes. In addition, ...
  199. [199]
    Human germline editing in the era of CRISPR-Cas
    Sep 11, 2020 · At present, CRISPR-Cas does not work sufficiently precise, leading to so-called off-target effects, i.e. unintended changes in non-target ...<|control11|><|separator|>
  200. [200]
    Chinese scientist who produced genetically altered babies ... - Science
    He Jiankui, the Chinese researcher who stunned the world last year by announcing he had helped produce genetically edited babies, has been found guilty of ...
  201. [201]
    CRISPR'd babies: human germline genome editing in the 'He ...
    The world was shocked in Nov. 25, 2018 by the revelation that He Jiankui had used clustered regularly interspaced short palindromic repeats ('CRISPR') to edit ...
  202. [202]
    Responding to claims of germline gene editing in humans
    In November 2018, the scientific community was rocked when He Jiankui, a Chinese biophysicist, claimed that he had overseen the birth of twin girls from ...
  203. [203]
    Human Germline and Heritable Genome Editing: The Global Policy ...
    Oct 20, 2020 · Seventy-five of the 96 countries prohibit the use of genetically modified in vitro embryos to initiate a pregnancy (heritable genome editing).
  204. [204]
    Study: There Is No Country Where Heritable Human Genome ...
    Oct 31, 2020 · In contrast, 78 out of 96 countries have policies to address heritable genome editing (i.e. for reproduction), where 70 prohibit this type of ...
  205. [205]
    The case for germline gene correction: state of the science
    The greatest clinical potential for heritable genome editing is in the prevention of monogenic diseases that so far have proven intractable to other therapeutic ...
  206. [206]
    Risks and benefits of human germline genome editing - NIH
    There is an ongoing debate whether genetic enhancement is ethically acceptable. This an issue that surely needs a focused analysis that cannot be given here.
  207. [207]
    Gene Editing and the Slippery Slope Argument: Should We Fix the ...
    Mar 21, 2018 · It seems that we are ultimately obliged (as is often the case) to trace a moral distinction on the therapy/enhancement dichotomy.
  208. [208]
    Human Gene Editing and the Ethics of Enhancement - ELSIhub
    Dec 6, 2022 · It also underlined the importance of the debates over enhancement, by raising the question of whether providing superior resistance to HIV ...
  209. [209]
    Debunking the slippery slope argument against human germ-line ...
    It argues that if genetic enhancement is governed by proper regulations and is accompanied by adequate education, then it need not violate recognized principles ...
  210. [210]
    Affordable Pricing of CRISPR Treatments is a Pressing Ethical ...
    Oct 18, 2024 · Casgevy, the world's first approved CRISPR-based cell therapy, has been priced at $2.2 million per patient. Although this hefty price tag ...
  211. [211]
    Gene Editing for Rare Genetic Diseases: Is An Equitable Future ...
    Mar 21, 2024 · Gene editing technologies such as CRISPR/Cas9 systems can be employed to offer targeted interventions and improve clinical outcomes.
  212. [212]
    Navigating equity in global access to genome therapy expanding ...
    Apr 4, 2024 · Issues of accessibility and affordability are pivotal in ensuring the equitable application of this novel therapy to enhance global health.
  213. [213]
    CRISPR therapies can treat disease but cost millions. An equity ...
    Oct 10, 2024 · CRISPR therapies can treat disease but cost millions. An equity-based approach could bring them to more people.
  214. [214]
    Towards affordable CRISPR genomic therapies: a task force ...
    Nov 8, 2023 · While pricing and access are concerns for genomic therapies here in the US and in other high income countries, these concerns are compounded ...<|separator|>
  215. [215]
    Human genome editing: a framework for governance
    Jul 12, 2021 · The Committee developed a governance framework that draws from good practices in the governance of emerging technologies and applied them ...
  216. [216]
    Human genome editing: recommendations
    Jul 12, 2021 · A set of two publications that provide advice and recommendations on appropriate institutional, national, regional and global governance mechanisms for human ...
  217. [217]
    Human Gene Therapy Products Incorporating Human Genome Editing
    Mar 5, 2024 · In this guidance, we, FDA, are providing recommendations to sponsors developing human gene therapy products incorporating genome editing (GE) of human somatic ...
  218. [218]
    United States: Germline / Embryonic
    Germline gene editing is banned in the United States by acts of Congress although there is no federal legislation that dictates protocols or restrictions.
  219. [219]
    European Union: Crops / Food - Global Gene Editing Regulation ...
    Gene editing and New Genomic Techniques are effectively banned for cultivation as they are regulated under the same strict regulations that sharply limit ...Missing: USDA 2023-2025
  220. [220]
    European Parliament votes to ease regulation of gene-edited crops
    Feb 7, 2024 · The European Parliament voted to lessen regulatory oversight of crops created through one type of DNA manipulation: gene editing.
  221. [221]
    All clinical research using germline genome editing banned in China
    Jul 19, 2024 · China's science and technology ministry has prohibited all clinical research involving germline genome editing, describing the approach as 'irresponsible'.
  222. [222]
    [PDF] Innovation and Interdependence: The Case of Gene-Editing ...
    First, lower barriers to entry create opportunities for forum-shopping by researchers, firms, and other actors. This facilitates regulatory arbitrage as actors ...
  223. [223]
    Full article: Risk-appropriate regulations for gene-editing technologies
    Jan 12, 2024 · Regulatory barriers lead to two key outcomes: reduced innovation investments and fewer commercialized products and technologies.
  224. [224]
    [PDF] List of barriers to implementation of genome editing technology in ...
    Based on these discussions some of the main barriers identified to uptake of the technology were regulatory and legislative ambiguity, societal perception, off- ...
  225. [225]
    Regulation as key to fulfilling the promises of agricultural genomics
    Jun 16, 2025 · This article provides a short review of recent scholarship on gene editing regulation and argues that regulation is an important part of the research ...