ABSTRACT
CRISPR‐Cas9 technology has rapidly advanced as a transformative genome‐editing platform, facilitating precise genetic modifications and expanding therapeutic opportunities across various diseases. This review explores recent developments and clinical translations of CRISPR applications in oncology, genetic and neurological disorders, infectious diseases, immunotherapy, diagnostics, and epigenome editing. CRISPR has notably progressed in oncology, where it enables the identification of novel cancer drivers, elucidation of resistance mechanisms, and improvement of immunotherapies through engineered T cells, including PD‐1 knockout CAR‐T cells. Clinical trials employing CRISPR‐edited cells are demonstrating promising results in hematologic malignancies and solid tumours. In genetic disorders, such as hemoglobinopathies and muscular dystrophies, CRISPR‐Cas9 alongside advanced editors like base and prime editors show significant potential for correcting pathogenic mutations. This potential was affirmed with the FDA's first approval of a CRISPR‐based therapy, Casgevy, for sickle cell disease in 2023. Neurological disorders, including Alzheimer's, ALS, and Huntington's disease, are increasingly targeted by CRISPR approaches for disease modelling and potential therapeutic intervention. In infectious diseases, CRISPR‐based diagnostics such as SHERLOCK and DETECTR provide rapid, sensitive nucleic acid detection, particularly valuable in pathogen outbreaks like SARS‐CoV‐2. Therapeutically, CRISPR systems target viral and bacterial genomes, offering novel treatment modalities. Additionally, CRISPR‐mediated epigenome editing enables precise regulation of gene expression, expanding therapeutic possibilities. Despite these advances, significant challenges remain, including off‐target effects, delivery methodologies, immune responses, and long‐term genomic safety concerns. Future improvements in editor precision, innovative delivery platforms, and enhanced safety assessments will be essential to fully integrate CRISPR‐based interventions into standard clinical practice, significantly advancing personalised medicine.
Keywords: cancers, CRISPR‐Cas systems, epigenome editing, genetic therapy, iPSC, molecular diagnostic techniques, nervous system diseases, personalised medicine
CRISPR‐Cas systems offer transformative genome editing capabilities for precise manipulation of cellular genes. This enables two main therapeutic avenues: ex vivo modification of patient cells for re‐transplantation or direct in vivo gene targeting via advanced delivery methods. These approaches unlock significant potential for developing targeted therapies across diverse diseases, advancing personalised medicine.

Abbreviations
- AAV
adeno‐associated virus
- ABE
adenine base editors
- ABL
abelson murine leukemia viral oncogene homologue 1
- AD
Alzheimer Disease
- AHC
adrenal hypoplasia congenital
- ALS
amyotrophic lateral sclerosis
- AML
acute myeloid leukemia
- APOBEC
apolipoprotein B mRNA editing catalytic polypeptide‐like
- APP
amyloid precursor protein
- ATTR
amyloid polyneuropathy, transthyretin‐related
- BCR
B cell receptor
- BE
base editors
- BLISS
breaks labelling in situ and sequencing
- BRAF
B‐Raf proto‐oncogene, serine/threonine kinase
- CAR
chimeric antigen receptor
- CasMINI
a compact, miniature CRISPR‐Cas system (e.g., a small Cas12f variant)
- CBE
cytosine base editors
- CF
cystic fibrosis
- CLAN
CRISPR loci annotation (i.e., a tool or nomenclature for annotating CRISPR loci)
- CLL
chronic lymphocytic leukaemia
- CML
chronic myeloid leukaemia
- CRC
colorectal cancer
- CRISPR
clustered regularly interspaced short palindromic repeats
- CVD
cardiovascular disease
- DETECTR
DNA endonuclease targeted CRISPR trans reporter
- DLBCL
diffuse large B‐cell lymphoma
- DMD
Duchenne muscular dystrophy
- DMT
DNA methyl transferase
- DREBIC
drug response evaluation by in vivo CRISPR
- DS
down syndrome
- DrS
Dravet syndrome
- DSB
double‐strand break
- EBV
Epstein–Barr virus
- EGFR
epidermal growth factor receptor
- EMT
epithelial‐to‐mesenchymal transition
- ES
embryonic stem
- EV
extracellular vesicles
- FA
Fanconi anaemia
- FDA
food and drug administration
- FL
follicular lymphoma
- FXS
fragile X syndrome
- HBB
haemoglobin beta
- HCC
hepatocellular carcinoma
- HD
huntington's disease
- HDR
homology‐directed repair
- HH
hypogonadotropic hypogonadism
- HHL
hereditary hearing loss
- HIV
human immunodeficiency virus
- HPV
human papillomavirus
- HR
homologous recombination
- HSC
haematopoietic stem cells
- HT1
hereditary tyrosinemia type I
- IE
immediate‐early
- KRAB
Krüppel‐associated box
- LHNP
lipid hybrid nanoparticles
- MCL
mantle cell lymphoma
- MEK
mitogen‐activated protein kinase kinase
- MeSH
medical subject headings
- MICA
MHC class I polypeptide‐related sequence A
- MMEJ
microhomology‐mediated end joining
- MND
motor neuron disease
- NHEJ
non‐homologous end joining
- NHGRI
national human genome research institute
- NK
natural killer
- OS
overall survival
- PAM
protospacer‐adjacent motif
- PARP
poly ADP‐ribose polymerase
- PD
Parkinson's disease
- PE
prime editors
- PKU
phenylketonuria
- PPC
patient‐derived primary cells
- PRISMA
preferred reporting items for systematic reviews and meta‐analyses
- PV
polycythemia vera
- RecQ
RecQ helicase
- RESCUE
RNA editing for specific c‐to‐u exchange
- RNA
ribonucleic acid
- RNP
ribonucleoprotein particle
- RP
retinitis pigmentosa
- RPA
replication protein A
- SCD
sickle cell disease
- SCID
severe combined immune deficiency
- SMA
spinal muscle atrophy
- SMN
survival‐motor neuron
- SSA
single strand annealing
- TALEN
transcription activator‐like effector nucleases
- TGFR
transforming growth factor beta receptor
- UCD
urea cycle disorder
- VEGFA
vascular endothelial growth factor A
- ZFN
zinc‐finger nucleases
1. Background
CRISPR was first discovered by Ishino et al., and later characterised by Doudna and Charpentier [1, 2]. The CRISPR sequences are regarded as the adaptive immune systems in many bacteria and archaea to protect them from bacteriophage, viral DNA, and plasmid invasion [3, 4]. The CRISPR‐Cas9 system consists of the Cas9 endonuclease and a guide RNA (gRNA) [1, 5], which scans the genome for a complementary sequence adjacent to a protospacer adjacent motif (PAM) [6, 7]. Once the PAM is recognised, Cas9 creates a double‐strand break (DSB) upstream of the PAM site [8, 9].
DNA damage activates repair pathways such as non‐homologous end joining (NHEJ) and homology‐directed repair (HDR) [2, 10] NHEJ, a template‐free mechanism, joins broken DNA ends, often resulting in small insertions or deletions that can cause gene knock‐outs. HDR, effective in the S and G2 phases, uses a homologous sequence for precise repair and allows for gene correction or knock‐ins [11, 12, 13, 14] Gene knock‐outs are more efficient due to the limited activation of HDR during certain cell cycle stages [15].
The CRISPR‐Cas system can precisely edit target DNA, offering potential treatments for genetic diseases and various refractory conditions, from sickle cell anaemia to cancer [16, 17]. Despite global research efforts, technical challenges in treating genetic diseases persist. This review examines the CRISPR‐Cas molecular mechanism, types, applications, and strategies in personalised medicine, along with associated challenges and limitations.
2. Method
2.1. Objective and Search Strategy
This study investigates CRISPR‐Cas technology's potential in treating challenging diseases by examining pre‐clinical and clinical studies to highlight its efficacy in managing difficult‐to‐treat disorders. A comprehensive literature search was conducted using databases such as PubMed, Web of Science, and Google Scholar. Keywords included ‘CRISPR Technology’, ‘Genetic Therapy’, ‘Clinical Applications’, ‘Delivery Methods’, ‘Monogenic disorders’, ‘Neurological disorders’, ‘Infectious diseases’, ‘Immunotherapy’, ‘Epigenetic editing’, The search covered articles published between 2010 and 2023.
2.2. Eligibility Criteria
English studies
Studies included in this review were original research articles, clinical trials, and review articles that focused on the application of CRISPR technology in treating diseases between 2010 and 2023.
Studies reporting CRISPR technology to treat challenging diseases, including cancers, neurodegenerative disorders, genetic diseases, infectious diseases, and autoimmune disorders
Studies report on the use of personalised medicine approaches for treating challenging diseases
Studies reporting on the clinical applications of CRISPR technology and personalised medicine in human patients
Studies reporting on the safety, efficacy, and feasibility of CRISPR technology and personalised medicine approaches
2.2.1. Exclusion Criteria
Non‐English studies
Studies outside the period between 2010 and 2023
Studies that did not report on the use of CRISPR technology or personalised medicine to treat challenging diseases
Studies that did not report the clinical applications of CRISPR technology or personalised medicine in human patients
Articles not available in English, studies not involving CRISPR‐Cas systems, and papers with insufficient methodological details
The authors assessed studies that were not peer reviewed or of low quality.
2.3. Study Selection
Two reviewers (M.A.H and M.A) conducted the study selection process independently according to the eligibility criteria. The screening process was performed by EndNote V.20 as follows: after yielding the initial articles, (1) the duplicate articles were removed, (2) the bi‐step title abstract process was done, and (3) a full‐text assessment was performed. Finally, studies that fully met the eligibility criteria were considered for data synthesis. If two reviewers disagree, a third senior reviewer (B.F.) makes the final decision.
3. Results
The search was conducted and yielded 4359 articles. After removing duplicates (n = 1453), 2906 articles were screened based on their title and abstract for relevance. Full‐text articles were then retrieved for the remaining 271 articles and assessed for eligibility according to the inclusion and exclusion criteria. Finally, 197 articles were included for data synthesis within the context. Figure 1 represents the PRISMA flowchart of the screening process. The included studies demonstrated the potential of CRISPR technology in various therapeutic areas. Key findings include successful applications in cancer immunotherapy, correction of genetic mutations in inherited diseases, and the development of CRISPR‐based diagnostics for infectious diseases. The majority of studies reported high specificity and efficacy of CRISPR systems, though challenges such as off‐target effects and delivery methods remain significant hurdles.
FIGURE 1.

PRISMA flowchart of the screening process.
3.1. Molecular Aspects of CRISPR
3.1.1. Molecular Mechanism of CRISPR Structure and Function
The CRISPR‐Cas9 system comprises two main components: a guide RNA (gRNA) and a CRISPR‐associated (Cas) nuclease. The gRNA is an RNA molecule that recognises and binds precisely to the target DNA, guiding the Cas nuclease to cut or edit [18, 19]. Ribonucleoprotein (RNP)‐mediated CRISPR genome editing, which involves the Cas9 protein and a single guide RNA (sgRNA), has advantages such as higher efficiency, improved cell viability, ease of transfer, and avoidance of plasmid‐related issues. The Cas nucleases are classified into Type 1 and Type 2, with six subdivisions [20] (Table 1). The gRNA is composed of two parts: the CRISPR RNA (crRNA), which contains a 17–20 nucleotide sequence that is complementary to the target DNA, and a tracer RNA that is 80 nucleotides long and binds to the crRNA, serving as a scaffold for the Cas nuclease [21]. The PAM is a short specific sequence located immediately after the complementary DNA sequence, and it is crucial for Cas nuclease cleavage. The PAM, approximately 2–6 nucleotides long, is positioned on the non‐target strand downstream of the targeted DNA sequence, directing the Cas nuclease to cleave 3–4 nucleotides upstream of it (Table 1) [7, 22].
TABLE 1.
General classification of CRISPR‐Cas system‐based Cas protein type, PAM site, and target sequence.
| Cas type | PAM site | Structure and signature | Target | |
|---|---|---|---|---|
| Cas protein Class 1 | Type I (subtype A–G) |
I‐A: 5‐CCN‐3 I‐B: 5‐TTC ACT‐3 5‐TAA −3 5‐TAT‐3 5‐TAG‐3 5‐ACA‐3 I‐C: 5‐NTTC‐3 I‐U: NA I‐D: NA I‐E: 5‐AAG‐3 I‐F: 5‐GCGC‐3 |
Multi subunit Cas protein; Signature protein: I: Cas3 |
Single strand DNA |
| Type III (subtype A–D) | Not recognised |
Multi subunit Cas protein; Signature protein: III: cas10 |
DNA/RNA | |
| Type IV (subtype A–C) | Not recognised |
Multi subunit Cas protein; Signature protein: IV: Csf1 |
Not–recognised | |
| Cas protein Class 2 | Type II (subtype A–C) |
5‐NGG‐3 |
Single subunit Signature protein: II: Cas9 |
DNA |
| Type V (subtype A–I, K) |
V‐A: 5‐TTTN‐3 V‐B: 5‐TTT‐3 5‐TTA‐3 5‐TTC‐3 V‐C & V‐D: 5‐AT rich PAM V‐E: 5‐TTCN‐3 V‐U 1 to 5 Not recognised |
Single subunit Signature protein: II: Cas12 Cas14(12f) |
Cas12: dsDNA/ssDNA Cas14: ssDNA |
|
| Type VI (subtype A–C) |
VI‐A: Non‐G (predicted) VI‐B1 & B2: NAN/NAA‐3 (Predicted) |
Signature protein: Signature protein: II: Cas13 |
Single strand RNA | |
| Type I (subtype A–G) |
I‐A: 5‐CCN‐3 I‐B: 5‐TTC ACT‐3 5‐TAA −3 5‐TAT‐3 5‐TAG‐3 5‐ACA‐3 I‐C: 5‐NTTC‐3 I‐U: NA I‐D: NA I‐E: 5‐AAG‐3 I‐F: 5‐GCGC‐3 |
Multi subunit Cas protein: Signature protein: I: Cas3 |
Single strand DNA |
Note: The CRISPR‐Cas system has revolutionised genetic engineering, allowing for precise and efficient manipulation of genetic material. The Cas proteins play a crucial role in this process by recognising and binding to specific DNA sequences, thereby facilitating targeted editing.
3.1.2. CRISPR‐Cas9 Mediated Repairing System
Unlike the effector complexes of Meganucleases, ZFNs, and TALENs, which rely on protein‐DNA interactions, the CRISPR‐Cas system leverages RNA–DNA base pairing and protein‐DNA interactions (Figure 2A) [23]. Furthermore, because of the long target sequence of CRISPR‐Cas (> 20 nucleotides) in comparison with Meganucleases, ZFNs, and TALENs (< 20 nucleotides), the CRISPR‐Cas system has been more precise and effective than others [21, 24]. Genome editing technologies that manipulate genome sequences take advantage of endonucleases to induce DS DNA DSBs at desired locations in the genome [25]. The processing of DSBs by the cellular DSB repair machinery is a determining factor in the type and success of gene manipulation outcomes. Therefore, the accuracy and efficiency of genome editing are influenced by the cellular DSB repair pathways [26]. Double‐strand breaks (DSBs) pose a threat to genome integrity, and cells have evolved multiple repair mechanisms. Therefore, the cellular DSB repair pathways influence the accuracy and efficiency of genome editing [27]. In Figure 2B, the CRISPR‐Cas9 system and the mechanisms of DSB repair are briefly depicted. The cell picks the particular repair pathway depending on different species, the cell cycle, target site sequence, and chromatin structure. Overall, mammalian cells intrinsically choose cNHEJ throughout the cell cycle, although HR and SSA are more frequently utilised during the S/G2 phase [28, 29].
FIGURE 2.

CRISPR‐Cas9, ZFN, and TALEN as genome editing tools. (A) CRISPR‐Cas9, through a single guide RNA complemented to target DNA and Cas9 protein creates a double‐stranded DNA cut. ZFN and TALEN‐engineered DNA‐binding proteins fused with the restriction enzyme FOK1 facilitate targeted genome editing by creating DNA double‐strand breaks. (B) The broken DNA will undergo the repair pathways: NHEJ or HDR; DSBs are repaired by NHEJ directly ligating the broken ends together with minimal DNA end processing when the ring‐shaped Ku70/Ku80 protein heterodimer binds to the DSB ends, which protects the DNA ends from exonucleases and also recruits the DNA‐dependent protein kinase and DNA ligase subsequently. Like NHEJ, MMEJ does not require a template for repairing the damaged DNA and begins with short‐range resection of the DSB. In MMEJ, DSB ends are re‐aligned using overhanged microhomologous sequences, and further the remaining 3’ ssDNA flaps are cleaved off, which results in small indels. ssDNA overhangs (> 1000 nucleotides) are generated by the 5′–3′ exonuclease activities of either EXO1 or BLM‐DNA2, and then coated by RPA, followed by the binding of RAD52, which mediates SSA pathway. Unless, RPA‐ssDNA can serve as a substrate for the RAD51 filament assembly, allowing for repair by HDR.
3.1.3. CRISPR Variants‐Based Target Modifications
DNA or RNA sequence modification based on CRISPR is performed using different enzymes such as Cas9, Cas12, and Cas14, and also the complexes of fused enzymes under the guidance of guide RNAs. Single‐strand or double‐strand DNA cleavage using these enzymes triggers repair processes, leading to gene disruption and indel mutations through NHEJ, or precise sequence modification can be achieved through HDR in the presence of template DNA [30, 31]. The fusion of Cas9 nickase (nCas9) or catalytically inactivated Cas9 (dead Cas9 or dCas9), which has been fused with functional domains of base editing enzymes, can also be used for genetic and epigenetic modifications, including base editing, to specific DNA sequences [32] (Figure 3). The base editors (BEs) comprise a cytidine deaminase like APOBEC mediated by dCas9, and nCas9 acts. This complex acts as an in situ modification, which in the case of dCas9 does not require the double‐strand breaks, while nCas9 is used to target specific loci through nick generation [33]. There are two prominent base editors: cytidine base editors (CBEs), which allow C to T conversions, and adenine base editors (ABEs), which enable A to G conversions. Additionally, the ADAR enzyme (Adenosine deaminases acting on RNA) fused with dCas13 for reversible and temporal A to G conversion (RNACas13‐ADAR) in fusions enables targeted editing of RNA [34, 35]. The transposase‐CRISPR‐mediated targeted integration can be induced by recruiting transposase complexes along with the guidance of catalytically dead Cas12k, resulting in targeted integration of gene fragments into the genome as a homology‐independent knock‐in strategy [36] (Figure 3).
FIGURE 3.

CRISPR‐Cas system strategies for gene manipulation via sequence editing, DNA integration, and transcriptional regulation. This figure illustrates multiple CRISPR‐based strategies for nucleotide and gene expression modification. Gene disruption or correction is achieved by CRISPR nucleases such as Cas9, Cas12, and Cas14, which introduce double‐strand breaks (DSBs) at target DNA sites, subsequently repaired by non‐homologous end joining (NHEJ) or homology‐directed repair (HDR). DNA and RNA base editing is performed using catalytically impaired Cas proteins (nCas9, nCas12, dCas13) fused to deaminase enzymes, allowing A‐to‐G or C‐to‐T conversions in DNA (by CBEs and ABEs) and reversible C‐to‐U or A‐to‐I conversions in RNA (via RESCUE or ADAR fusion systems). Targeted transposon integration is enabled by Cas12k in complex with transposase components such as TnsB, TnsC, and TniQ. Transcriptional regulation is mediated by dCas9 fused to transcriptional effectors such as KRAB (repression, CRISPRi) or VP16/VP64 (activation, CRISPRa), modulating gene expression at the promoter level. Finally, epigenetic modulation is accomplished by fusing dCas9 to chromatin‐modifying enzymes including histone acetyltransferases (HATs), deacetylases (HDACs), DNA methyltransferases (DMTs), and histone methylation/demethylation regulators (HMTs, HDMs), enabling locus‐specific and heritable gene expression changes.
dCas13 has been used for a wider variety of transcriptome regulation applications in addition to its well‐established function in ADAR‐mediated A‐to‐I RNA editing. For example, the RESCUE system (RNA Editing for Specific C‐to‐U Exchange) couples dCas13 with a cytidine deaminase domain to convert cytidine (C) to uridine (U) in target RNAs, thereby expanding the RNA editing toolkit beyond A‐to‐I modifications [37]. Furthermore, non‐coding RNAs, such as long non‐coding RNAs and microRNAs, can be functionally regulated using dCas13‐based platforms that target RNA molecules without altering the underlying genomic DNA [38]. Catalytically inactive Cas13 (dCas13) can be directed to specific RNA transcripts to modulate gene expression post‐transcriptionally—either by repressing translation through steric hindrance of ribosome binding or by enhancing expression via fusion to translation initiation factors [39, 40]. This RNA‐level regulation is transient and reversible, offering a significantly lower risk of long‐term off‐target effects compared to DNA editing methods, making it an attractive strategy for therapeutic and research applications [38] (Figure 3).
Prime Editing is a genome editing technique that enables precise DNA insertions, base substitutions, and specific deletions using a mechanism that functions without DSBs and donor templates [41]. Prime Editing achieves its operations by integrating Cas9 nickase and reverse transcriptase elements into one protein complex that utilises pegRNA for direct DNA modification guidance [41, 42]. By extending editing operations and shielding the genome from unnecessary mutations, prime editing produces a precise tool with adaptable genome modification capabilities that can be used for a range of therapeutic purposes(Figure 3).
The CRISPRa and CRISPRi variants can either stimulate or inhibit gene expression, depending on the attached regulatory region. The dCas9‐KRAB complex mediates transcriptional repression of genes within their endogenous genomic loci, while the dCas9‐VP64version of CRISPRa was developed by fusing four VP16 units (VP64) and regarded as a solid transcriptional activator [43, 44]. Site‐specific gene regulation can be achieved by altering DNA structure or histone modifications. CRISPR‐mediated epigenetic editing has shed light on allele‐specific epigenome editing, which utilises inactive dCas9 as a DNA‐binding domain and fused enzymes such as DNA methyl transferase (DMT), histone methyl transferase(HMT), histone demethylase(HDM), acetyltransferase(HAT), and deacetylases(HDAC) that can be targeted to alter the epigenetic state at precise locations within the genome [45, 46] (Figure 3).
3.1.4. CRISPR‐Mediated off‐Targets
CRISPR‐Cas systems can bind to unintended genomic sites and cleave DNA, leading to off‐target effects [47, 48]. These off‐target events can cause large deletions, genomic rearrangements, and lethal mutations that result in loss of gene function and may promote malignant transformation [49, 50]. Efforts to mitigate off‐target effects include enhancing on‐target efficiency and employing in silico tools for off‐target detection.
Off‐target effects occur when CRISPR mediates the excision of DNA sequences that are similar, but not identical, to the intended target, often due to sequence mismatches. It is crucial to recognise these potential risks and the ongoing strategies to minimise them [51]. Several advancements, including truncated guide RNAs (gRNAs), dual sgRNA strategies (using the nickase platform), high‐fidelity Cas9 variants, and CRISPR base editors (BE), have been developed to enhance specificity and reduce off‐target cleavage [52]. Various techniques are available for detecting off‐target effects, such as BLISS (in situ detection), LAM‐PCR HTGTS (translocation sequencing), DISCOVER‐Seq (ChIP‐based detection), GUIDE‐seq (anchored primer enrichment), CIRCLE‐seq (in vitro selection libraries), and Digenome‐seq and SITE‐Seq (in vitro genomic DNA digestion) [53, 54]. Unbiased and biased methods, such as Elevation and other detection techniques, are commonly used to identify off‐target mutations. Moreover, newly engineered Cas variants with enhanced precision have demonstrated significant reductions in off‐target activity [52].
3.2. Applications of CRISPR: Delivery Methods
Along with genome editing tools, delivery methods have been developed recently. CRISPR‐Cas9 delivery methods involve two main categories: viral and non‐viral [55]. The delivery vehicle will determine what types of DNA, mRNA, or protein can be delivered and what delivery methods achieve a safe and efficient in vivo/ex vivo delivery system for CRISPR‐Cas9 [56]. Efficient delivery of CRISPR components is critical for the success of gene editing therapies. Viral vectors such as adeno‐associated virus (AAV) and lentivirus have been widely used because of their high transduction efficiencies; however, concerns regarding immunogenicity, limited packaging capacity, and potential insertional mutagenesis remain significant challenges [55]. As depicted in Figure 4A, non‐viral delivery systems are broadly classified into physical methods (e.g., microinjection, electroporation, hydrodynamic delivery, gene gun, sonoporation, magnetofection), polymer‐based nanoparticles (e.g., polyplexes, gold and inorganic nanoparticles, dendrimers), lipid‐based nanoparticles (e.g., LNPs, liposomes, lipoplexes), and biological‐biochemical methods (e.g., exosomes, MENDs, antibody‐ and receptor‐mediated delivery, CPPs, ITOP). These platforms offer safer and more flexible alternatives to viral vectors, with reduced immunogenicity; however, their efficiency and target specificity remain ongoing challenges. Figure 4A and Table S1 [56, 57].
FIGURE 4.

Clinical strategies and delivery platforms for CRISPR‐Cas‐based gene therapy. (A) This figure summarises key clinical applications and delivery methods for CRISPR‐Cas systems in therapeutic development. Delivery approaches are categorised into viral and non‐viral systems. Viral vectors—including lentivirus, adenovirus, adeno‐associated virus (AAV), herpes simplex virus (HSV), and baculovirus—are commonly used for their high transduction efficiency and long‐term gene expression capabilities, though each carries unique limitations in immunogenicity, cargo capacity, and integration risk. Non‐viral delivery systems are subdivided into physical (e.g., microinjection, electroporation, nucleofection, gene gun, sonoporation, hydrodynamic delivery), chemical (e.g., lipid nanoparticles, lipoplexes, gold nanoparticles, dendrimers, DNA nanoclews, inorganic nanoparticles), and biological/biochemical approaches (e.g., cell‐penetrating peptides, iTOP, receptor‐mediated endocytosis, exosomes, antibody‐based targeting). These strategies enable the delivery of CRISPR cargo in the form of plasmid DNA, mRNA, or ribonucleoprotein (RNP) complexes. The figure highlights how optimising delivery platforms is essential to overcome barriers in targeting efficiency, immune response, and in vivo applicability for clinical translation of CRISPR‐based therapies. (B) In Vivo Gene Therapy: CRISPR‐Cas systems are delivered directly into the patient using viral vectors (e.g., AAV, Lentivirus, Retrovirus) for localised (e.g., muscle) or systemic (e.g., liver, lung) gene editing. Non‐viral methods employing nanoparticles (DNA, organic, inorganic, MSN, MEND) and cell‐penetrating peptides also facilitate direct in‐body delivery. Ex Vivo Gene Therapy: Patient cells (e.g., HSCs, T‐cells, iPSC‐derived NK cells) are collected, genetically modified with CRISPR‐Cas systems in vitro, selected for desired modifications, and then re‐infused back into the patient for therapeutic effect.
3.3. CRISPR Therapeutics: Bridging Ex Vivo Engineering and in Vivo Precision
The therapeutic strategy of CRISPR‐Cas systems could be categorised into ex vivo and in vivo. Recent advancements in engineered nanoparticles and targeted delivery strategies have improved the in vivo performance of non‐viral systems, thereby enhancing the therapeutic potential of CRISPR‐based interventions. This in‐depth understanding and optimisation of delivery methods are essential for translating CRISPR technology into clinical practice [58]. The ex vivo strategy consists of extracting and isolating targeted cells from a patient, editing them, and, as autologous transplantation, delivering the cells back to the patient [59, 60]. The summarised schematic of viral and non‐viral delivery methods is depicted in Ex vivo strategies deliver important advantages from controlled cell manipulation as well as superior quality control capabilities and the ability to increase modified cell numbers before re‐administration while presenting difficulties in achieving effective cell engraftment as well as logistical complexities during transplantation processes. The clinical advantage of direct patient delivery for CRISPR components through in vivo approaches includes quicker therapeutic action, although challenges exist regarding delivery efficiency along with immune responses and off‐target effects control. The analysis of these methods reveals their distinct advantages and disadvantages for researchers to plan upcoming discussions about particular in vivo uses [61]. In vivo, strategy is conducted by direct delivery of disease‐specific designed CRISPR‐Cas vectors to target cells or organs of the body as in situ gene therapy [57] (Figure 4B).
CRISPR‐Cas9 is being used ex vivo for cancer immunotherapy, specifically in constructing CAR‐T and TCR‐T cells [62, 63] and treating hereditary diseases such as sickle cell anaemia, β‐thalassaemia, etc. [64, 65]. Also, viral infection elimination, which is extensively and effectively used against human immunodeficiency virus (HIV), hepatitis B virus [66], and human papillomavirus (HPV) [67, 68] are engineered T cells designed to target cells for improved treatment outcomes. The main clinical application based on an in vivo strategy is monogenic disorders arising from a single‐gene defect inherited according to traditional Mendelian patterns [69]. Emerging new era treatment approaches using CRISPR to edit patient‐derived iPSCs (CRISPR iPS cells) enable scientists to correct disease‐causing mutations and a broad range of challenging diseases like cellular immunotherapies for cancer treatment and replace them by re‐introducing back into patients [70, 71]. Although the generation and initial gene editing of iPSCs are performed in vitro, the overall process is considered an ex vivo approach because the modified cells are subsequently expanded, quality‐controlled, and re‐transplanted into the patient [70] (Figure 4B).
iPSC therapy‐based treatment is classified into three main strategies: the first one is gene knock‐out, which is mainly applied to eliminate viral infections or any approach that mediates the elimination of the disease‐causing gene [72]; the second is gene knock‐in, which is an exogenous nucleotide sequence introduced to CRISPR iPS to correct a mutated gene or replace the mutated gene with the correct version of a gene/specific sequence [73]. Transcription regulation is regarded as a third strategy, which mediates transcriptional activator or suppressor of a candidate endogenous genes as a therapeutic strategy when increased or decreased expression of endogenous genes causes disease [74] (Figure 4B).
3.4. Miniaturised CRISPR Tools for Increased Clinical Applicability
Recent advancements in CRISPR technology have prioritised the miniaturisation of CRISPR systems to improve clinical translation by enhancing in vivo delivery and reducing immunogenicity. Compact nucleases such as Cas12f (CasMINI), CasX (also known as Cas12e), and Casθ have been engineered for their smaller size and efficient activity, making them more suitable for packaging into delivery vectors like AAVs. Researchers have improved editing performance by optimising guide RNAs—such as introducing circular guide RNAs (cgRNAs) for stability—and fusing effector domains to enable potent transcriptional regulation. CasX and Casθ, derived from non‐pathogenic bacteria, also show promising specificity and low off‐target activity, further enhancing their therapeutic potential. These developments aim to overcome the limitations of traditional CRISPR‐Cas9 systems and support broader applications in treating genetic disorders [75, 76, 77, 78].
3.5. Application of CRISPR in Clinic
CRISPR technology has implications for several challenging diseases, such as cancer, genetic disorders, neurologic disorders, and infectious diseases, regarding diagnosis and treatment. CRISPR‐Cas9 genome editing has been extensively investigated in preclinical and clinical settings [79].
3.5.1. CRISPR Applications in Cancer: Gene Correction, Onco‐Immunotherapy, and Beyond
Cancer is a multidimensional situation comprised of environmental and intrinsic factors. Genomic mutations as a main clue in cancer pathobiology may occur as a new gene mutation, an inherited allele, or a mutation that the repair system fails to correct [80]. In any case, it causes the activation of oncogenes and/or inactivation of tumour suppressors, leading to disturbance in gene expression, the epigenome, metabolism, and eventually disruption of cell proliferation, structure, polarity, and motility [81]. In addition, such a complex situation is made by evading and circumventing the host's defence systems [82]. Gaining insights into the intricate coordination of molecular and cellular events and the consequent modifications in the tumour microenvironment is paramount in comprehending the onset, advancement, and response to therapies in diverse cancer types [83]. Such understanding serves as a cornerstone in developing innovative and more efficacious treatment approaches, with the ultimate goal of enhancing outcomes for the large number of individuals diagnosed with cancer each year [84]. It seems necessary to explore and develop a strategy to target and correct mutated genes by directly eliminating, restoring, correcting, or repairing mutated sequences/genes (Table 2) [105]. Also, an effective treatment strategy should be considered according to gene mutations and their subsequent effect on the epigenetic imprint, gene expression (based on functional mechanisms of mutated or epigenetics), corresponding protein, and molecular treatment efficacy and application [106].
TABLE 2.
A comprehensive overview of the various approaches of treatment utilising CRISPR in cancer therapy.
| Author date | Approaches | Cancer type | Main findings | Ref |
|---|---|---|---|---|
| Rosenblum, Gutkin et al. 2020 |
CRISPR Cas9 genome editing using targeted lipid nanoparticles |
Glioblastoma and ovarian tumours |
|
[85] |
| Chen, Liu et al. 2017 |
The CRISPR‐Cas9 system, which uses clustered regularly interspaced short palindromic repeats |
Glioma |
|
[86] |
|
Zhang, Zhang et al. 2017 |
Delivering a CRISPR‐Cas9 system usinPLNP‐basedPLNP based approach |
Malignant melanoma, prostate cancer, and breast cancer |
|
[87] |
| Behan, Iorio et al. 2018 |
Exploiting the stress phenotype caused by genetic changes such tumour or suppressor gene loss by the use of medications that target the oncogene or the activated downstream signalling pathway |
NA |
|
[88] |
| Meca‐Cortés, Guerra‐Rebollo et al. 2017 |
Therapeutic cells and GCV |
Glioblastoma |
|
[89] |
| Liang, Li et al. 2017 |
CRISPR‐Cas9 genome editing technology, VEGFA targeting |
Osteosarcoma |
|
[90] |
| Wang, Chow et al. 2019 |
CRISPRa mediated MAEGI |
Pancreatic cancer |
|
[91] |
| Tang and Shrager, 2016 |
Surgery, radiation therapy, and/or targeted/chemotherapy |
Lung cancer |
|
[92] |
| Zhang, Wu et al. 2020 |
Gene therapy and chemotherapy |
Hepatoma carcinoma (HCC) |
|
[93] |
| Zhang, Li et al. 2018 |
Inhibitors of B Raf proto‐oncogene, serine/threonine kinase (BRAF), inhibitors of EGFR |
Melanoma, non‐small cell lung cancer |
|
[94] |
|
Rayner, During et al. 2019 |
RNP and plasmid‐based methods |
Colorectal cancer (CRC) |
|
[95] |
| KouranovaEvguenia, ForbesKevin et al. 2016 |
Column chromatography, precipitation with ethanol, and purification with 5 M ammonium sulfate all use the same volume of starting material |
Glioma |
|
[96] |
| Deng, Tan et al. 2019 |
In vivo delivery of the CRISPR Cas9 genome editing method to suppress tumour cell PD‐L1 production by knocking out the Cyclin‐dependent kinase 5 (Cdk5) gene |
Murine melanoma and triple‐negative breast cancer |
|
[97] |
| Szlachta, Kuscu et al. 2018 |
Chemotherapy and pharmacologic inhibition of MEK signalling inhibitors |
Pancreatic ductal adenocarcinoma (PDAC) |
|
[98] |
| Lin, Larrue et al. 2020 | NA |
Acute myeloid leukaemia (AML) |
|
[99] |
| Liu, Zhao et al. 2018 |
Gene delivery of CRISPR‐Cas9 system with nanocarriers |
Chronic myeloid leukaemia (CML) |
|
[100] |
| Cheung, Chow et al. 2018 |
Antibody‐based targeted therapy and synthetic lethality |
Lung cancer |
|
[101] |
| Wang and Sun 2017 |
CRISPR‐Cas9‐mediated targeting of the oncogene HER2 |
Breast cancer |
|
[102] |
| Ibrahim, Özsöz et al. 2019 |
Editing, modifying, deleting, adding, and replacing DNA sequences |
NA |
|
[103] |
| Cook and Ventura 2019 |
Immuno oncology therapeutic approaches involving engineered cell‐based therapy using CRISPR‐based approaches |
Non‐small cell lung cancer |
|
[104] |
Note: The table covers the cancer type that each approach is targeting, as well as the main findings of each study. Cancer therapy has been an area of intense research, and CRISPR technology has emerged as a promising tool for precision cancer treatment.
The concern of gene mutation behind cancer pathobiology caused the emergence of CRISPR‐Cas9 as a powerful method for making desired changes and corrections [107, 108]. CRISPR‐Cas9‐mediated established cells and animal models have resulted in clinical trials, which potentially treat tumours and offer great promise for inhibiting migration, invasion, and even treatment of tumours [107, 109, 110]. Some of the most clinically advanced strategies built from CRISPR systems present themselves in the form of immunotherapies which utilise CAR‐T and CAT‐T‐cell therapies. This method utilises CRISPR technology to improve cancer‐targeting functions in immune cells while providing additional therapeutic possibilities to direct gene editing [63]. CRISPR‐based CAR‐T therapies involve engineering T cells to express synthetic receptors targeting tumour antigens, often incorporating gene edits like PD‐1 or TCR knockouts to enhance efficacy and reduce exhaustion. In contrast, CRISPR‐altered T‐cell (CAT‐T) therapies focus on modifying endogenous T cells for CAT‐T therapies typically involves using gene editing tools like CRISPR‐Cas9 to alter their natural T‐cell receptors (TCRs). This process aims to redirect the T cells' inherent ability to recognise specific cancer antigens, often those presented by human leukocyte antigen (HLA) molecules, including patient‐specific neoantigens. The modification can involve inserting new TCR alpha and beta chains that confer desired specificity, which then complex with the existing CD3 components to form a functional, tumour‐targeting TCR [111, 112]. While CAT‐T approaches may offer improved safety profiles, their reliance on native TCR specificity can limit efficacy, particularly in tumours with low immunogenicity. Both strategies leverage CRISPR's precision to optimise therapeutic outcomes, with CAR‐T therapies demonstrating significant success in hematologic malignancies, and CAT‐T approaches being explored for solid tumours and other applications. Ongoing research continues to refine these modalities, aiming to balance efficacy, safety, and accessibility in cancer immunotherapy [107, 113]. The main approaches to treatment using CRISPR in cancer therapy studies are represented in Table 2 and schematically in Figure 5A.
FIGURE 5.

CRISPR‐Cas applications in clinical disease management and epigenetic regulation. This figure highlights the diverse and expanding clinical applications of CRISPR‐Cas technologies across five major therapeutic areas. In the cancer section, CRISPR is used to disrupt oncogenes, enhance immunotherapy (e.g., editing PD‐1 or TCRs), perform functional genomic screens to identify vulnerabilities, and support precision medicine strategies. In immunotherapy, CRISPR enables engineered T cells, CAR‐T cell modification, and checkpoint inhibition to improve immune targeting of tumours. The epigenome editing section illustrates CRISPR‐based gene regulation techniques such as CRISPR interference (CRISPRi) and activation (CRISPRa), targeted methylation via DNMT3A, and histone modifications through fusion with epigenetic enzymes. Precise transcriptional modulation is achieved without altering the DNA sequence. The histone methylation/demethylation area includes Cas‐fusion systems that specifically target histone markers, enabling dynamic chromatin remodelling. In infectious disease applications, CRISPR systems are employed for pathogen genome editing, antiviral strategies, and diagnostics using SHERLOCK/DETECTR platforms. Additionally, the lower portion of the figure illustrates emerging genome editing strategies such as prime editing and base editing, showing how cytosine and adenine base editors, along with reverse transcriptase‐guided pegRNAs, enable single‐nucleotide modifications without DSBs. This multi‐tiered representation underscores CRISPR's therapeutic versatility from genomic modification to epigenetic tuning and pathogen detection.
3.5.2. CRISPR‐Enhanced Immunotherapy: Engineering T Cells and NK Cells for Precise Medicine
Chimeric Antigen Receptor (CAR) T‐cell therapy, a ‘living drug’ using genetically engineered patient T cells to target and eliminate cancer, has revolutionised blood cancer treatment [114]. CRISPR‐Cas9 significantly enhances CAR‐T outcomes by enabling precise CAR gene integration, mitigating T‐cell exhaustion through knockout of inhibitory genes (e.g., PD‐1, CTLA‐4, and LAG‐3), and improving CAR‐T survival and anti‐cancer effects by interfering with programmed cell death [115, 116, 117, 118]. While T‐cell exhaustion remains a challenge, immune checkpoint inhibitors do not perpetually resolve CAR‐T drawbacks. CRISPR also facilitates “off‐the‐shelf” allogeneic CAR‐T development by removing endogenous TCRs to prevent GVHD and deleting MHC molecules to avoid host rejection [115, 116, 117, 119].
Besides disrupting inhibitory receptor removal, CRISPR‐Cas9 can interfere with the interaction between Fas and Fas ligands responsible for programmed cell death, increasing CAR‐T cells' anti‐cancer effect. Importantly, CRISPR‐Cas9 has a signature in removing the CAR‐T cell resistance [118]. The successful development of iPSC‐derived CAR‐engineered T (iCART) cells capable of eliminating cancer cells represents a pivotal advancement in clinical translation and therapeutic application (Figure 5D) [120].
CRISPR‐Cas9 has played a pivotal role in increasing the efficacy of NK (natural killer) cells in immunotherapy against AML [120]. NK cells, which are significant lymphocytes in the innate immune system, can be engineered using CRISPR‐Cas9 to knock out immune checkpoints and inhibitory signals, integrate tissue homing receptors to improve tumour infiltration, provide extra activating signalling to enhance anti‐tumour activity, and arm with chimeric antigen receptors (CARs) to enhance tumour specificity. CRISPR‐Cas9 can also be used to transcriptionally activate genes, such as the NKG2D ligand MICA, to improve the activation of NK cells [121]. In addition, CRISPR‐Cas9‐mediated disruption of PD‐1 and targeting of other immunosuppressive pathways, such as the adenosine 2A receptor, has been shown to enhance the efficacy of CAR‐T (chimeric antigen receptor‐T cell) therapy against cancer [122]. Furthermore, CRISPR‐Cas9‐mediated targeting of the Cish gene has been shown to enhance the cytotoxicity of primary human NK cells towards B lymphoma cells, demonstrating the potential of CRISPR in improving NK cell‐based immunotherapies [123, 124].
Modified NK cells generated using CRISPR‐Cas9 can express various receptors, such as high‐affinity DC16 Fc receptor or CAR receptors, which can improve their potency against solid tumours when combined with other therapies [125, 126]. Studies have shown the feasibility and efficacy of using induced pluripotent stem cells (iPSCs) as a platform to generate NK cells carrying cancer‐homing CAR receptors (CARiPSC‐NK cells), which demonstrated impressive anti‐tumour activity in ovarian cancer xenograft models with improved cell survival and expansion in vivo and less toxicity [127]. The FDA recently approved using off‐the‐shelf NK cells produced from clonal master iPSCs, named FT500, which synergized with T cells to treat solid tumour malignancies more effectively [128, 129] (Figure 5D).
For autoimmune conditions like systemic lupus erythematosus, cellular immunotherapies such as CD19‐targeted Chimeric Antigen Receptor (CAR) T cells are being explored to redirect immune cells and correct underlying autoimmunity by selectively eliminating pathogenic B cells, aiming for potential single‐administration cures [130, 131]. Building on these advancements, precision engineering of T cells, notably through CRISPR‐based chimeric autoantigen‐T cell receptors (CATCR), offers an even more refined approach. CATCR‐T cells are designed to reprogram a patient's T cells to specifically seek out and bind to receptors on autoimmune disease‐causing B cells, leading to their selective elimination while safeguarding normal immune function [132]. This innovative strategy provides tailored treatments that can control autoimmunity without the broad infection risks associated with conventional immunosuppressive therapies, marking a significant step towards bespoke solutions for devastating autoimmune conditions [133].
To ensure reproducibility and translational potential, studies utilising human stem cell‐derived immunotherapies, such as iPSC‐derived NK or CAR‐T cells, should adhere to the International Society for Stem Cell Research (ISSCR) guidelines. These standards, endorsed by journals such as Cell Proliferation, provide comprehensive recommendations for the characterisation, quality control, and ethical use of human pluripotent and tissue stem cells in research and clinical applications [134, 135].
3.5.3. Targeting Monogenic Diseases With CRISPR: From Bench to FDA‐Approved Therapies
The CRISPR‐based gene treatments for monogenic disorders have shown major clinical advancements that demonstrate their power for transformation. The breakthrough achievement in gene therapy emerged when the Food and Drug Administration (FDA) approved CRISPR‐based treatment CTX001 to treat sickle cell disease in the United States. The CRISPR‐based therapy CTX001 changes patient‐specific haematopoietic stem cells in a laboratory for fetal haemoglobin reactivation to solve disease symptoms while making clinical problems less severe [136]. Clinicians are currently studying CRISPR‐based therapeutic methods for genetic (inherited) diseases beyond sickle cell and beta‐thalassaemia as specific cases to prove the far‐reaching medical possibilities of this technology for treating genetic‐based disorders (Table 3).
TABLE 3.
An overview of the various approaches of treatment utilising CRISPR in Common Genetic Disorders.
| Disease | Monogenic or polygenic/target gene | Stem cell | Cell line | In vivo model | Transfer model | Ref |
|---|---|---|---|---|---|---|
| SCA | Monogenic/β‐globin | hiPSCs | HEK293T, bc1, TNC1 | Human | Electroporation, transfection | [137] |
| PV | Monogenic/JAK2 | hiPSCs | HEK293T | Human | Transfection | [138] |
| β‐ Thalassaemia | Monogenic/HBB gene | hiPSCs | Human |
Transfection |
[139] | |
| Cataracts | Monogenic/Crygc | SSCs | Mouse | Microinjection, transfection, electroporation | [140] | |
| A1AT deficiency | Monogenic/SERPINA1 | hiPSCs | HEK293T | Human | Transfection | [138] |
| FA | Monogenic/FANCC | Patient fibroblasts | Human | Transfection | [141] | |
| PKU | Monogenic/PAH | c.1222C>T COS‐7 | Human |
Transfection |
[142] | |
| CF | Monogenic/CFTR | Intestine Stem cell | HEK293T | Human | Transfection | [143] |
| UCD | Monogenic/OTC | MC57G | Mouse | Transfection | [144] | |
|
AHC and HH |
Monogenic/DAX1 | Monkey |
Microinjection |
[145] | ||
| HT‐I | Monogenic/Fah | 3 T3 cells | Mouse | Hydrodynamic injection | [146] | |
| Osteosarcoma | Monogenic/CDK11 | KHOS and U‐20S | Human |
Transfection, electroporation |
[147] | |
| CRC | Polygenic/APC, SMAD4, TP53, KRAS, PIK3CA | Human intestinal epithelial organoids | Human | Transfection | [148] | |
| Bronchial alveolar adenoma | Polygenic/Kras, p53, Lkb1 | mESCs | Neuro‐2a | Mouse |
Transfection |
[149] |
| CVD | Monogenic/Pcsk9 | 3 T3‐L1 | Mouse | Transfection | [150] | |
| Barth syndrome | Monogenic/TAZ | hiPSCs | PGP1 cell line | Human | Transfection | [151] |
| Intestinal hyperplasia | Monogenic/Apc | mESCs | Mouse | Blastocycst injection | [152] | |
| Cardiomyopathy | Monogenic/Myh6 | 10 T1/2 cells | Mouse | Intraperitoneal injection | [153] | |
| DS | Monogenic/GATA 1 | K562 | Human |
Transfection |
[154] | |
| RP | Monogenic/RGPR | iPSCs | Human | Transfection | [155] | |
| Contextual fear memory | Monogenic/Mecp2 |
HEK293FT |
Mouse |
Transfection Stereotactical injection |
[156] | |
| DMD |
Exon 45 of dystrophin gene, Exon 23 of dystrophin gene (Monogenic) |
iPSC |
Myoblasts, Muscle stem cells, Immortalised patient myoblasts, HEK293T, C2C12 and mdx |
Mice, Human | Microinjection, nanoparticle, transfection, electroporation, lipofection | [157, 158, 159, 160, 161, 162] |
| ALS and/or FTD | Polygenic/SOD, FUS, CHMP2B, Ku80, C9ORF72 | iPSC | LCL, FUS/TLS cell line | Human, G93A‐SOD1 transgenic mice | Electroporation, nucleofection, Adeno‐associated virus, transfection | [163, 164, 165, 166, 167, 168] |
| Dravet Sundrome | Monogenic/Scnla | Mouse embryonic hippocampal neurons | P‐19 | Mice | Adeno‐associated virus | [169] |
|
SCID X‐1 |
Monogenic/IL2Rg |
hESCs | K‐562, T hCD4+cells | Human |
Transfection |
[170] |
Abbreviations: AD, Alzheimer disease; AHC, Adrenal hypoplasia congenital; CF, cystic fibrosis; CRC, colorectal cancer; CVD, cardiovascular disease; DMD, Duchenne muscular dystrophy; DS, Down syndrome; DrS, Dravet syndrome; FA, Fanconi anaemia; HD, Parkinson disease; HH, hypogonadotropic hypogonadism; HT1, hereditary tyrosinemia type I; PD, Parkinson Disease; PKU; PhenylKetonUria; PV, Polycythemia Vera; RP, retinitis pigmentosa; SCA, sickle cell anaemia; SCID, severe combined immune deficiency; UCD, urea cycle disorder.
Clinical genetic disorder management is divided into two main approaches: pre‐ and post‐genomic. In pre‐genomic procedures, single‐base gene editing mediated by CRISPR is a hallmark, while when an original gene is damaged and inactivated, the inactivation of a single deleterious gene cannot address the complex situation of multifactorial disease [125]. Therefore, newly developed single‐base substitution tools, such as cytosine BE, ABE, and prime editors (PEs), could partially compensate for the limitations. In the era of post‐genomics, CRISPR‐Cas9 technology has been employed to develop new versions of chimer Cas protein to explore intended genes in genome splicing [171], transcription [172], modification [173], and epigenetic regulation [174] (Figure 5B).
3.5.4. CRISPR Against Infectious Diseases: Diagnostic and Therapeutic Frontiers
HIV, Hepatitis B, human papillomavirus, and recently, COVID‐19 are among the more challenging viruses, increasing healthcare concerns and socioeconomic burdens. Due to immune evasion, mutation, latency, and chronic infection leading to organ damage, fighting against some types of viruses is more challenging for new medical strategies. In this regard, CRISPR technology, as a unique gene‐editing tool, offers a novel medical approach to diagnosing and predicting threats of infectious diseases. The two analogous diagnostic systems, DETECTR [175, 176] and SHERLOCK [177, 178], have been designed to detect RNA and DNA sequences, respectively. Following the outbreak of COVID‐19 infection, besides real‐time polymerase chain reaction (RT‐PCR) and the Loop‐mediated isothermal amplification (LAMP) method, which are the most applied diagnostic methods for detecting the virus RNA [179]. Also, CRISPR‐based detection methods rapidly emerged as highly sensitive alternatives to RT‐PCR and LAMP, enabling cost‐effective and rapid diagnoses of SARS‐CoV‐2 [180, 181]. Additionally, CRISPR‐based therapeutic approaches have been developed to selectively target and eradicate viral and bacterial genomes [182] (Figure 5E).
CRISPR‐Cas9 is employed to target essential viral genes such as the long terminal repeats, CCR5, and CXCR4 of the HIV virus, utilising single or dual‐guide RNAs to enhance editing efficiency [183]. Targeting viral replication factors like BST‐2/tetherin and promoter regions has further improved viral suppression [184, 185, 186, 187, 188, 189, 190]. Similarly, CRISPR‐Cas9 has demonstrated efficacy in disrupting the oncogenes E6 and E7 of high‐risk HPV types (HPV‐16 and HPV‐18) [191] leading to cell growth inhibition and apoptosis induction [192] with ongoing research extending to low‐risk HPV types (e.g., HPV‐6/11) and vaccine development [193]. Moreover, CRISPR‐Cas13, targeting RNA, has been investigated for its antiviral activity against SARS‐CoV‐2, effectively inhibiting viral replication in human respiratory cells [194]. Several studies were conducted to explore the anti‐COVID‐19 effect of CRISPR. It is documented that the CRISPR‐ased antiviral modality potently inhibits viral replication and activity in human respiratory epithelial cells and cleaves 100% of positive RNA viruses [195] (Table 4).
TABLE 4.
An overview of the various approaches of treatment utilising CRISPR in common viral disease.
| Disease | Target gene | Stem cell | Cell line | In vivo model | Transfer model | References |
|---|---|---|---|---|---|---|
| HIV‐1 resistance | CCR5 | hiPSCs | HEK293T | Human | Transfection | [196] |
| HIV‐1 infection and immunisation |
LTR loci of integrated viral genome, T10 |
J‐lat | CHME5 | Human | Transfection | [197] |
| EBV |
Multiple –gene EBV nuclear antigen 1 (EBNA‐1) |
Ranji and Burkitt's lymphoma cell lines |
Human | Transfection | [198] | |
| HPV and cervical cancer | HPV16, E7 oncogenes | SiHa and Caski | Human | Transfection | [199] | |
| HBV | p53 and Pten gene | Huh7, HepG2 | Mouse |
Transfection, electroporation hydrodynamic Injection |
[200, 201] | |
| Cryptosporidiosis | C. parvum | HTC8 | Mouse | Transfection | [202] | |
| Herpes Simplex 1 (HSV‐1) | Multiple genes |
Vero cells human oligodendroglioma cells |
Human | Transfection | [203] | |
| Human herpesvirus 6 (HHV‐6) | Eradication the viral genome trough targeting conserved region of the DRs |
293 T cell line harbouring the integrated virus genome (293 T‐6A) & iciHHV‐6A patient cells |
Human | Transfection | [204] | |
| Human cytomegalovirus (CMV) | Immediate‐early (IE) genes |
HFF primary fibroblasts THP‐1 monocytic cell line |
Human | Transfection | [205] | |
| JC polyomavirus | JCV T‐antigen | HJC‐2 cells | Human | Transfection | [206] | |
| SARS‐CoV‐2 |
RdRP / N gene regions |
Cas13d A549 cells huMAN cells (PAC‐MAN) |
Human | Transfection | [195] |
Abbreviations: DrS, Dravet syndrome; EBV, Epstein–Barr virus; HBV, hepatitis B virus; HIV, human immunodeficiency virus; HPV, human papillomavirus.
Besides HIV and HPV infection, CRISPR was recruited to eliminate other well‐known DNA‐based viruses, including Epstein–Barr virus (EBV), cytomegalovirus, Sars‐cov‐2, JC virus, and Zika virus [207, 208] (Table 4). Although CRISPR technology is derived from bacteria, it has been used against bacterial infections such as Acinetobacter baumannii, Escherichia coli , and Staphylococcus aureus ‐induced osteomyelitis by eliminating infectious agents and resolving drug resistance [209, 210, 211].
3.5.5. Application of CRISPR in Neurological Disorders
Genetic mutations are responsible for several neurological diseases, and gene editing approaches are currently used to diminish the burden of neurological disease [212]. Although disease‐modifying therapies were approved for some neurological disorders, there is a lack of disease‐modifying therapies for some other conditions, and clinicians have tried symptom therapy [213]. Although CRISPR‐Cas9 was applied to set up neurological disease animal models, CRISPR targeting neurological disorder strategies is currently used to treat neurological diseases in the clinical setting (Figure 5B) [214] (Table 5). The initial CRISPR‐Cas9 application in neurological disease was presented in the monogenic condition, where CRISPR was used in Duchenne muscular dystrophy (DMD) [227]. Subsequent studies focused on other monogenic disorders of the nervous system, such as spinal muscular atrophy (SMA), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and Alzheimer's disease (AD) in both in vitro and in vivo settings [228]. Moreover, CRISPR can also be applied to hereditary neurological disorders like Down syndrome, fragile X syndrome, Tay‐Sachs, Sandhoff, and Niemann‐Pick, well‐known as incurable diseases (Table 5).
TABLE 5.
A comprehensive overview of the various approaches of treatment utilising CRISPR in neurological disorders.
| Disease (abbreviations) | Target gene | Delivery method & model | Stem cell | Cell line | Animal model | References |
|---|---|---|---|---|---|---|
| AD | APP, PSEN2, Gmf, Bacel | Transfection, Adeno‐associated virus, nucleofection, nanocomplexes | Fibroblast, iPSCs |
Primary cortical neurons and hippocampus of Tg2576 mice, BV2 cells, CA3 hippocampus |
AD patients, mice | [215, 216, 217] |
| ALS and/or FTD | SOD, FUS, CHMP2B, Ku80, C9ORF72 | Electroporation, nucleofection, Adeno‐associated virus, transfection | iPSC | G93A‐SOD1, C9ORF72‐ALS/FTD | Human, mice | [163, 164, 165, 166, 167, 168] |
| Ds | Scnla | Adeno‐associated virus | DS mice | [169] | ||
| Epilepsy | Kcnal (Monogenic) | Adeno‐associated virus | Camk2a‐Cre mice and epileptic mice | [218] | ||
| HD | HTT gene (Monogenic) | Stereotactic injection, lentivirus, Adeno‐associated virus | BM‐MSCs | HEK293T | Mouse |
, [157] |
| PD |
SNCA, hM4Di, hM3Dq, mcu, sRAGE, DJ1, parkin, PINK1, LRRK2 (Polygenic) |
Lentivirus, Adeno‐associated virus, microinjection | iPSCS, UCB‐MSCs, MD NPCs, zebrafish, Procrine fetal fibroblasts | Human, zebrafish, pig | [223, 224, 225] | |
|
Dyslexia |
DIP2A (Monogenic) |
PGK‐Puro‐P2A‐mCherry | homozygous knockout 46C ESC cell line | Murine ES cell | [226] |
3.5.6. Prospects of CRISPR‐Mediated Epigenetic Therapeutic Interferences
Epigenetic modifications of genomic DNA and histone proteins, such as methylation or acetylation, at specific genomic loci or histone residues, have been shown to play critical roles in biological fates.
The emergence of the CRISPR‐Cas9 system led to access to DNA and histones for epigenetic editing. Utilising HNH/UvC Dead Cas9 as a DNA‐binding domain, fused to enzymes such as DNA methylases, histone acetyltransferases/deacetylases, and histone methyltransferases/demethylases can be targeted to alter the epigenetic state at precise locations (Figure 5C) [174].
Recruitment of epigenome editing effector domains utilising CRISPR‐Cas systems allows for the stimulation and repression of endogenous gene expression and graded control of gene regulation [229]. Fusing chromatin‐modifying domains enables targeted epigenome editing of nuclease‐deactivated Cas9 (dCas9) in cultured cells and animal models. On the other hand, delivering large dCas9 fusion proteins to desired tissues and target cells is an obstacle for in vivo studies.
For example, a nuclease‐dead Cas9 fused to the p300 histone acetyltransferase domain (dCas9–p300) has been shown to target H3K27 acetylation at specific loci, driving robust transcriptional activation; in transgenic mouse models, gRNA‐targeted dCas9–p300 altered epigenetic states and downstream gene expression in the brain and liver in vivo [230]. Notably, burn injury activates a histone H3.1 Ser10 phosphorylation signal in spinal dynorphinergic (Pdyn) neurons, and CRISPR‐Cas9‐mediated S10 → A mutagenesis of H3.1 in these neurons (via AAV delivery) blocked this modification and significantly elevated thermal nociceptive thresholds in mice [231]. Complementary CRISPR modalities also enable targeted genetic modulation in tissue trauma: base editors allow precise single‐base corrections without double‐strand breaks, and dCas9–KRAB or Cas9 nuclease strategies can respectively repress or knock out genes, facilitating gene silencing or knock‐in approaches for pain and tissue‐damage pathways (Figure 5C) [232].
3.5.7. Current Landscape of CRISPR‐Based Therapeutics in Clinical Applications: Recent Approvals and Ongoing Trials
CRISPR gene editing has reached significant clinical milestones as of 2025, and several promising treatments have emerged from trials. The first CRISPR–Cas9–based cell treatment for sickle cell disease (SCD) was exagamglogene autotemcel (Casgevy), which was licenced in late 2023 [233]. In early 2024, it was approved for transfusion‐dependent β thalassaemia [234]. This ex vivo therapy edits a patient's haematopoietic stem cells to induce fetal haemoglobin, and 94% of treated SCD patients achieved freedom from vaso‐occlusive crises for at least 1 year [233]. Beyond haemoglobin disorders, in vivo CRISPR therapies are advancing rapidly. Transthyretin amyloid cardiomyopathy (ATTR‐CM) can be treated with NTLA‐2001, a one‐time intravenous CRISPR treatment that has advanced to a pivotal Phase 3 trial after showing > 90% reduction in pathogenic TTR protein levels in patients [235] and clinical metrics indicating disease stabilisation. Similar to this, NTLA‐2002, an in vivo CRISPR therapy for hereditary angioedema, reduced attack rates by 91%–97% with a single dosage [236] enabling patients to remain attack‐free, with no side effects reported, paving the way for a Phase 3 study.
CRISPR‐engineered cell therapies are also showing promise in oncology. In its Phase 1 trial, a CRISPR‐edited allogeneic CAR T‐cell treatment (CB‐010) for recurrent B‐cell lymphoma produced a 94% overall response rate, including 69% total complete remissions [237]. The first‐ever durable full remission (lasting more than 3 years) in solid tumours was achieved with an off‐the‐shelf CAR‐T method using CRISPR‐edited T cell therapy (CTX130) that targets CD70 in advanced renal cell carcinoma [238]. This result highlights the potential for gene‐edited immune cells to tackle solid cancers, and next‐generation versions (e.g., CTX131) are already under development. Beyond traditional CRISPR nucleases, base‐editing approaches have entered the clinic: a base editor therapy (VERVE‐101) for heterozygous familial hypercholesterolemia demonstrated substantial LDL cholesterol reductions (50% lower sustained at 6 months) after a one‐time treatment [239], marking the first proof‐of‐concept for base editing in humans. Even infectious diseases are being targeted—a CRISPR‐based therapy aimed at curing HIV (EBT‐101) has shown that the approach is feasible and safe in early trials, although viral rebound occurred after stopping antiretroviral therapy (8), underscoring the challenges ahead.
This finding underscores how gene‐edited immune cells can take on solid cancers, and next‐generation versions (e.g., CTX131) are in the pipeline. In addition to conventional CRISPR nucleases, base‐editing strategies have now been brought into the clinic: a base editing therapy (VERVE‐101) for heterozygous familial hypercholesterolemia showed robust reductions in LDL cholesterol (50% decrease at 6 months after a single treatment) [239], representing the first proof‐of‐principle for base editing in humans. Even infectious diseases are being targeted—a CRISPR‐based therapy for the cure of HIV (EBT‐101) has been shown to be both feasible and safe in early trials, but has resulted in viral rebound following discontinuation of ART [240] Several clinical trials have explored the impact of CRISPR in patients with cancer. This article mentions the recent (from 2018) clinical trials documented at ClinicalTrials.gov in Table S2.
3.5.8. Diagnosis
Beyond its therapeutic potential, CRISPR‐Cas9 holds significant promise in diagnostics, offering rapid, sensitive, specific, accurate, and cost‐effective detection for pathogens, early cancer diagnosis, single nucleotide polymorphisms, and genetic diseases. Notable platforms include SHERLOCK (Specific High‐sensitivity Enzymatic Reporter unlocking), utilising Cas13 for highly sensitive and specific RNA detection in viral infections (e.g., Zika, dengue) and cancer‐related mutations [241, 242, 243]. DETECTR (DNA Endonuclease‐Targeted CRISPR Trans Reporter) is another CRISPR‐based diagnostic technique that uses the Cas12 enzyme to detect specific DNA sequences. DETECTR is a particular and sensitive tool for detecting mutations associated with genetic diseases, such as sickle cell anaemia, and identifying specific strains of bacteria [244]. These CRISPR‐based diagnostics offer a powerful alternative to traditional cancer screening methods, which often lack the necessary sensitivity and speed for early detection, potentially improving patient prognoses [245]. In colorectal cancer (CRC), where early detection significantly improves outcomes, CRISPR‐Cas9 shows promise in identifying key gene mutations (e.g., PIC3C3A, KRAS, TP53, SMAD4, APC) in CRC organoids and detecting de novo driver genes. Furthermore, CRISPR‐Cas13 technology offers a superior alternative to qRT‐PCR for detecting tumour‐derived microRNAs (e.g., miR‐23a, miR‐1290, miR‐126, and miR‐940), overcoming the limitations of traditional methods for early CRC diagnosis [246].
Besides its use in early diagnosis of CRC, CRISPR‐Cas9 can be applied as a screening modality to determine de novo diagnostic biomarkers like proteins, genes, and receptors involved in the cell signalling of pancreatic cancer cells, including drug sensitivity, apoptosis, proliferation, and spheroid formation [247]. For example, the overexpression of the HDAC1 gene induces epithelial‐to‐mesenchymal transition (EMT), a well‐established mechanism contributing to chemoresistance [248]. Similarly, the PRMT5 gene has been implicated in the development of chemoresistance; conversely, its inhibition renders pancreatic cancer cells more susceptible to gemcitabine, a commonly used first‐ or second‐line chemotherapeutic agent [249]. Approximately 130 genes are responsible for platinum resistance in pancreatic cancer cells [250]. The CRISPR‐Cas system represents a transformative approach for both curing and diagnosing pancreatic cancer, particularly in overcoming platinum resistance [251, 252]. Previous research reveals that CRISPR/Cas9 screening effectively identifies key genes, such as BRIP1, ERCC4, and FANCD2, alongside pathways enriched in DNA binding and NADH dehydrogenase activity, which modulate sensitivity to platinum‐based chemotherapies [251, 253]. By precisely editing these resistance‐conferring genes, CRISPR can synergistically enhance the efficacy of chemotherapy, for example, by disrupting oncogenic KRAS mutations or restoring tumour suppressor functions like TP53 [254]. In diagnostics, CRISPR‐based platforms like SPEAR offer highly sensitive and rapid detection of critical cancer‐related mutations, such as KRAS, from liquid biopsies, enabling earlier diagnosis and disease monitoring [255]. Despite its immense promise, challenges remain, notably the delivery of CRISPR components through the dense desmoplastic stroma of pancreatic tumours, although strategies like iRGD‐guided tumour‐penetrating nanocomplexes and stroma‐modulating agents are being explored to overcome this barrier [256, 257]. This integration of CRISPR‐mediated gene modification with personalised screening using patient‐derived organoids is paving the way for tailored, more effective treatment strategies against this aggressive cancer.
CRISPR can be utilised as a powerful detector of specific bacterial pathogens since CRISPR is one of the bacterial defence systems [258, 259]. Still, the diagnostic properties are not limited to only diagnosing pathogenetic bacteria. CRISPR can also be utilised to diagnose viral pathogens. The gold standard for diagnosing SARS‐CoV‐2 during the recent pandemic is the reverse transcription‐quantitative polymerase chain reaction (RT‐qPCR). However, this method has significant drawbacks, including its confinement to specialised laboratories, the requirement for skilled workers, and the possibility of erroneous or misleading data [260]. CRISPR‐based next‐generation diagnostic technologies offer a solution to these issues; they are affordable, sensitive, specific, and do not necessitate complex equipment so that they may be employed in settings outside of the clinical field [261].
3.5.9. Challenges and Limitations CRISPR Bench to Bedside
Technology like the CRISPR‐Cas9 platform allows reverse mutagenesis, which could be used to treat disease [262]. To effectively manipulate the genome to remove and replace (Knock‐in) causative mutations or generate host alterations via insertion or deletion as protective functions is what is meant by the term ‘targeted gene therapy’ [58]. First, HDR to NHEJ must be overcome to replace a gene sequence with a provided fragment encoding the necessary repair to generate protein variants with healthy phenotypes. In the latter case, NHEJ is a crucial part of the HIV ongoing evaluation because it effectively generates a nonfunctional allele of the gene [55, 262]. Despite the treatment strategy, the complexity of the gene therapy candidate disease is another challenge in finding effective treatments [263]. In contrast to monogenic disorders, which can be easily treated by correcting a faulty copy of the culprit gene, polygenic diseases, and cancers require numerous simultaneous modifications to the genome to accomplish a successful treatment [69]. Another important challenge in gene therapy is that efficient and effective correction occurs at all cell cycle stages [264]. The efficacy of gene therapy depends on several factors, including the disease's genetic background and topologies of the target and pattern sequences, the type of therapeutic alteration, the repair mechanism intended to be recruited, the effectiveness of delivery techniques, and the cell state [265]. Therefore, before moving on to human trials, all therapeutic ideas must be proved in model organisms to address these problems and propose solutions [266]. All these challenges can be different from one person to another, which is the main description of personalised medicine. Even though the preceding explanation suggests that CRISPR‐Cas9 is a practical method, this gene editing system has various limitations that make its application in clinical trials problematic due to its recent identification and use in patients. A few approaches have been explored to address the CRISPR‐Cas9 system's drawbacks, including immunogenicity, off‐targeting, polymorphism, delivery technique, and ethics [17].
Some challenges prevent the widespread utilisation of CRISPR‐Cas9 in cancer research and therapies, the most significant of which is the technique's limited editing efficacy in tumours and the potential toxicity of existing delivery vehicles. To successfully implement genome editing systems in the intended species or cells, CRISPR‐Cas9 delivery relies on the availability of adequate and effective alternatives of delivery mechanisms. Unfortunately, getting the Cas9 system into living organisms has been difficult until now. The Cas9‐based gene editing platform has been delivered by integrating physical approaches with viral vectors. In vitro, the physical methods are more practical than the viral vector‐based methods, which often have low packing capacities and a high risk of side effects. Non‐viral drug delivery technologies, including polymeric and lipid nanocarriers, have shown promising delivery performance and targeting efficacy in recent preclinical and animal investigations to deliver CRISPR‐Cas9 systems. These adeno‐associated viruses are regarded as possible gene editing system transporters. The efforts to modify charged nanocarriers by altering their structures are described, and prospective non‐viral vectors undergoing clinical studies are highlighted. Delivering CRISPR‐Cas molecules into target cells is essential for therapeutic purposes, which have been facing difficulties since transfer vesicles can be affected by immune reactions, toxicity, or rapid degradation. Extracellular vesicles (EVs), a wide array of membrane vesicles, have been proposed as a hypothetical replacement. Many cells can generate EVs; exosomes are the smallest, with only 40–15 nm diameters, and are released by endocytes [267, 268]. Exosomes are carriers of molecules such as messenger RNA (mRNA) and miRNA, which can be absorbed by target cells through various forms of endocytosis or fusion with the plasma membrane [269]. This natural affinity of exosomes provides a significant opportunity to act as carriers of intercellular nucleic acids and therapeutic molecules, thus bypassing the aforementioned difficulties in transferring CRISPR‐Cas molecules into targets through biological membranes [270]. The CRISPR‐Cas9 technology, which can be directly applied to embryos, requires less time to implement compared to gene‐targeting technologies that use embryonic stem cells (ES cells). Very robust processes that guarantee the introduction of the intended mutation are now possible, thanks to advances in bioinformatics tools (to find the most acceptable sequences to build guide RNAs) and in optimising experimental settings.
DNA fragments (such as cDNAs) can be inserted into a genome using Cas9 to generate a DSB and activate the DNA repair machinery. Targeted alleles often have additional alterations, such as deletions, partial or multiple incorporations of the targeted vectors, or even duplications, because the DNA repair system's objective is not to insert DNA pieces into the genome. Even while secondary unwanted mutational events at the target locus plague conventional ES cell‐based experiments, researchers have found a means to avoid creating mice with passenger mutations. Most research facilities employ positive and negative selection procedures, as well as validation methods that seek out further mutations in the target location, to isolate ES cells that have undergone the desired recombination events. However, the CRISPR‐Cas9 strategy cannot select the intended goal when applied to embryos, greatly reducing the likelihood of identifying the target allele. Because of the observed mosaicism in CRISPR‐Cas9‐created founder mice, it is especially challenging to discover unwanted genomic modifications around the targeted region [271].
4. Closures and Conclusion
CRISPR‐Cas9 genome editing technology created a fundamental transformation in medical science that enables DNA modification through site‐specific programs with extraordinary precision and minimal difficulty. The clinical and research applications of CRISPR systems expand quickly but multiple essential obstacles need resolution to achieve their therapeutic maximum. The priority now is to boost CRISPR platform accuracy since minimal off‐target events can generate major implications during medical applications. Different upcoming genome editing tools including Cas12a, Cas13, base editors and prime editors demonstrate potential to enhance both specificity and genome editing capability. Predictive algorithms together with machine learning‐guided gRNA design techniques provide essential tools to decrease off‐target activities. The deployment of genome editing remains the main bottleneck that stands in the way of its use for translation. The refinement of both viral vectors as well as non‐viral delivery vehicles (lipid nanoparticles and polymeric systems) continues but tissue‐specific scalable delivery systems functioning in a transient manner represent the upcoming priority. The combination of nanotechnology and synthetic biology has demonstrated potential to create specialised solutions for dealing with these problems. Research demonstrates that CRISPR shows rising applicability in treating complex diseases like malignancies in addition to neurodegenerative conditions and autoimmune disorders and monogenic disorders. New therapeutic opportunities emerge for polygenic diseases by implementing combination gene editing approaches along with epigenetic therapy methods such as dCas9‐p300 and dCas9‐KRAB and CRISPR‐based on/off control tools. Social and ethical along with regulatory issues possess similar importance to the field. The development and implementation of socially responsible policies require interdisciplinary teams to address problems related to germline editing and CRISPR‐based therapy availability as well as long‐term medical safety. Visual engagement between stakeholders becomes essential to guarantee proper clinical implementation with ethical standards. Future progress in CRISPR depend on combining it with AI‐powered target identification systems which pair with high‐throughput single‐cell screening methods and intelligent delivery platforms to become a fundamental precision medicine platform. The next 10 years will determine whether CRISPR advances from groundbreaking technology into the everyday medical practice for curing difficult‐to‐treat diseases.
Author Contributions
Bahareh Farasati Far and Sherko Nasseri contributed to the conception and design of the study. Material preparation and data collection were conducted by Sherko Nasseri, Mohammad Amin Habibi, and Bahareh Farasati Far. All authors participated in drafting the initial version of the manuscript. Sherko Nasseri redesigned and rewrote the manuscript, including the design of five figures in alignment with the revised content. During the revision process, Bahareh Farasati Far, Marziyeh Akbari, Morteza Katavand, and Sherko Nasseri contributed to all manuscript modifications. All authors reviewed and approved the final version of the manuscript. Sherko Nasseri supervised all steps of manuscript preparation, revision, and submission.
Ethics Statement
The authors have nothing to report.
Consent
The authors have nothing to report.
Conflicts of Interest
The authors declare no conflicts of interest.
Supporting information
Table S1. Overview of viral and non‐viral CRISPR delivery methods and their characteristics. This table summarises a broad range of non‐viral delivery systems developed for transporting CRISPR components—including DNA, RNA, and ribonucleoprotein (RNP) complexes—into target cells. The methods are categorised by their underlying mechanisms (physical or chemical), and each entry details the delivery approach, cargo type, major advantages, and associated drawbacks. Technologies include well‐established platforms like electroporation and lipofection, as well as emerging nanotechnologies such as gold nanoparticles, DNA nanoclews, and multifunctional envelope‐type nano devices (MENDs). The table emphasises each method’s utility for in vitro or in vivo applications, editing efficiency, cytotoxicity, delivery specificity, and translational feasibility. This comparative overview supports the selection of optimal non‐viral delivery strategies in CRISPR‐based research and therapeutic development.
Table S2. Ongoing clinical trials and applications of CRISPR. Summary of significant clinical developments and recent studies using CRISPR‐based therapies. As of early 2025, this table lists notable instances of CRISPR technology applications through clinical trials. In a variety of disease areas, such as genetic disorders, oncology, cardiovascular disease, and infectious disease, it highlights a range of therapeutic approaches, such as ex vivo cell therapies, in vivo gene editing via nucleases base editors, and engineered cell therapies. Key reported discoveries or outcomes, the target indication, the current clinical trial phase, the particular therapy or application, and pertinent citations are all included in the details.
Acknowledgements
To improve the language quality of the manuscript, the authors used AI‐assisted editing tools for grammar correction, sentence restructuring, and clarity enhancement.
Far B. F., Akbari M., Habibi M. A., Katavand M., and Nasseri S., “ CRISPR Technology in Disease Management: An Updated Review of Clinical Translation and Therapeutic Potential,” Cell Proliferation 58, no. 11 (2025): e70099, 10.1111/cpr.70099.
Funding: The authors received no specific funding for this work.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.
References
- 1. Deshpande K., Vyas A., Balakrishnan A., and Vyas D., “Clustered Regularly Interspaced Short Palindromic Repeats/Cas9 Genetic Engineering: Robotic Genetic Surgery,” American Journal of Robotic Surgery 2, no. 1 (2015): 49–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Doudna J. A. and Charpentier E., “Genome Editing. The New Frontier of Genome Engineering With CRISPR‐Cas9,” Science 346, no. 6213 (2014): 1258096. [DOI] [PubMed] [Google Scholar]
- 3. Rath D., Amlinger L., Rath A., and Lundgren M., “The CRISPR‐Cas Immune System: Biology, Mechanisms and Applications,” Biochimie 117 (2015): 119–128. [DOI] [PubMed] [Google Scholar]
- 4. Barrangou R. and Marraffini L. A., “CRISPR‐Cas Systems: Prokaryotes Upgrade to Adaptive Immunity,” Molecular Cell 54, no. 2 (2014): 234–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Asmamaw M. and Zawdie B., “Mechanism and Applications of CRISPR/Cas‐9‐Mediated Genome Editing,” Biologics 15 (2021): 353–361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Chen H., Fu Y., Feng K., et al., “Polydopamine‐Coated UiO‐66 Nanoparticles Loaded With Perfluorotributylamine/Tirapazamine for Hypoxia‐Activated Osteosarcoma Therapy,” Journal of Nanobiotechnology 19, no. 1 (2021): 298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Gleditzsch D., Pausch P., Müller‐Esparza H., et al., “PAM Identification by CRISPR‐Cas Effector Complexes: Diversified Mechanisms and Structures,” RNA Biology 16, no. 4 (2019): 504–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Jiang F. and Doudna J. A., “CRISPR‐Cas9 Structures and Mechanisms,” Annual Review of Biophysics 46, no. 1 (2017): 505–529. [DOI] [PubMed] [Google Scholar]
- 9. Doudna J., “CRISPR Book Review—Doudna Responds,” Nature 547, no. 7662 (2017): 162. [Google Scholar]
- 10. Yang Y., Xu J., Ge S., and Lai L., “CRISPR/Cas: Advances, Limitations, and Applications for Precision Cancer Research,” Frontiers in Medicine 8 (2021): 649896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Liu M., Zhang W., Xin C., et al., “Global Detection of DNA Repair Outcomes Induced by CRISPR‐Cas9,” Nucleic Acids Research 49, no. 15 (2021): 8732–8742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Hille F. and Charpentier E., “CRISPR‐Cas: Biology, Mechanisms and Relevance,” Philosophical Transactions of the Royal Society, B: Biological Sciences 371, no. 1707 (2016): 20150496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Nambiar T. S., Baudrier L., Billon P., and Ciccia A., “CRISPR‐Based Genome Editing Through the Lens of DNA Repair,” Molecular Cell 82, no. 2 (2022): 348–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lieber M. R., “The Mechanism of Double‐Strand DNA Break Repair by the Nonhomologous DNA End‐Joining Pathway,” Annual Review of Biochemistry 79 (2010): 181–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Lau C. H., Tin C., and Suh Y., “CRISPR‐Based Strategies for Targeted Transgene Knock‐In and Gene Correction,” Faculty Reviews 9 (2020): 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Wang S.‐W., Gao C., Zheng Y. M., et al., “Current Applications and Future Perspective of CRISPR/Cas9 Gene Editing in Cancer,” Molecular Cancer 21, no. 1 (2022): 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Rasul M. F., Hussen B. M., Salihi A., et al., “Strategies to Overcome the Main Challenges of the Use of CRISPR/Cas9 as a Replacement for Cancer Therapy,” Molecular Cancer 21, no. 1 (2022): 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Doudna J. A. and Charpentier E., “The New Frontier of Genome Engineering With CRISPR‐Cas9,” Science 346, no. 6213 (2014): 1258096. [DOI] [PubMed] [Google Scholar]
- 19. Wu X., Kriz A. J., and Sharp P. A., “Target Specificity of the CRISPR‐Cas9 System,” Quantitative Biology 2, no. 2 (2014): 59–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Liu Z., Dong H., Cui Y., Cong L., and Zhang D., “Application of Different Types of CRISPR/Cas‐Based Systems in Bacteria,” Microbial Cell Factories 19, no. 1 (2020): 172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Zhang H. X., Zhang Y., and Yin H., “Genome Editing With mRNA Encoding ZFN, TALEN, and Cas9,” Molecular Therapy 27, no. 4 (2019): 735–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Schubert M. S., Thommandru B., Woodley J., et al., “Optimized Design Parameters for CRISPR Cas9 and Cas12a Homology‐Directed Repair,” Scientific Reports 11, no. 1 (2021): 19482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Castro N. G., Bjelic J., Malhotra G., et al., “Comparison of the Feasibility, Efficiency, and Safety of Genome Editing Technologies,” International Journal of Molecular Sciences 22, no. 19 (2021): 10355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Li H., Yang Y., Hong W., Huang M., Wu M., and Zhao X., “Applications of Genome Editing Technology in the Targeted Therapy of Human Diseases: Mechanisms, Advances and Prospects,” Signal Transduction and Targeted Therapy 5, no. 1 (2020): 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Xue C. and Greene E. C., “DNA Repair Pathway Choices in CRISPR‐Cas9‐Mediated Genome Editing,” Trends in Genetics 37, no. 7 (2021): 639–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Hisano Y., Sakuma T., Nakade S., et al., “Precise In‐Frame Integration of Exogenous DNA Mediated by CRISPR/Cas9 System in Zebrafish,” Scientific Reports 5 (2015): 8841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Zhang J. P., Li X. L., Li G. H., et al., “Efficient Precise Knockin With a Double Cut HDR Donor After CRISPR/Cas9‐Mediated Double‐Stranded DNA Cleavage,” Genome Biology 18, no. 1 (2017): 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Yoshimi K., Kunihiro Y., Kaneko T., Nagahora H., Voigt B., and Mashimo T., “ssODN‐Mediated Knock‐In With CRISPR‐Cas for Large Genomic Regions in Zygotes,” Nature Communications 7 (2016): 10431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Kosicki M., Tomberg K., and Bradley A., “Repair of Double‐Strand Breaks Induced by CRISPR‐Cas9 Leads to Large Deletions and Complex Rearrangements,” Nature Biotechnology 36, no. 8 (2018): 765–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Huang J., Zhou Y., Li J., Lu A., and Liang C., “CRISPR/Cas Systems: Delivery and Application in Gene Therapy,” Frontiers in Bioengineering and Biotechnology 10 (2022): 942325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zhang M., Eshraghian E. A., Jammal O. A., Zhang Z., and Zhu X., “CRISPR Technology: The Engine That Drives Cancer Therapy,” Biomedicine & Pharmacotherapy 133 (2021): 111007. [DOI] [PubMed] [Google Scholar]
- 32. Karlson C. K. S., Mohd‐Noor S. N., Nolte N., and Tan B. C., “CRISPR/dCas9‐Based Systems: Mechanisms and Applications in Plant Sciences,” Plants (Basel) 10, no. 10 (2021): 102055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Huang L., Yang C., Chen Y., Deng H., Liao Z., and Xiao H., “CRISPR‐Mediated Base Editing: Promises and Challenges for a Viable Oncotherapy Strategy,” Human Gene Therapy 34, no. 15–16 (2023): 669–681. [DOI] [PubMed] [Google Scholar]
- 34. Porto E. M., Komor A. C., Slaymaker I. M., and Yeo G. W., “Base Editing: Advances and Therapeutic Opportunities,” Nature Reviews. Drug Discovery 19, no. 12 (2020): 839–859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Marina R. J., Brannan K. W., Dong K. D., Yee B. A., and Yeo G. W., “Evaluation of Engineered CRISPR‐Cas‐Mediated Systems for Site‐Specific RNA Editing,” Cell Reports 33, no. 5 (2020): 108350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Rezazade Bazaz M., Ghahramani Seno M. M., and Dehghani H., “Transposase‐CRISPR Mediated Targeted Integration (TransCRISTI) in the Human Genome,” Scientific Reports 12, no. 1 (2022): 3390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Rees H. A. and Liu D. R., “Base Editing: Precision Chemistry on the Genome and Transcriptome of Living Cells,” Nature Reviews Genetics 19, no. 12 (2018): 770–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cox D. B. T., Gootenberg J. S., Abudayy O. O., et al., “RNA Editing With CRISPR‐Cas13,” Science 358, no. 6366 (2017): 1019–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Apostolopoulos A., Kawamoto N., Chow S. Y. A., et al., “dCas13‐Mediated Translational Repression for Accurate Gene Silencing in Mammalian Cells,” Nature Communications 15, no. 1 (2024): 2205, 10.1038/s41467-024-46412-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Abudayyeh O. O., Gootenberg J. S., Konermann S., et al., “C2c2 Is a Single‐Component Programmable RNA‐Guided RNA‐Targeting CRISPR Effector,” Science 353, no. 6299 (2016): aaf5573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Anzalone A. V., Randolph P. B., Davis J. R., et al., “Search‐and‐Replace Genome Editing Without Double‐Strand Breaks or Donor DNA,” Nature 576, no. 7785 (2019): 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lu C., Kuang J., Shao T., et al., “Prime Editing: An All‐Rounder for Genome Editing,” International Journal of Molecular Sciences 23, no. 17 (2022): 9862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Omachi K. and Miner J. H., “Comparative Analysis of dCas9‐VP64 Variants and Multiplexed Guide RNAs Mediating CRISPR Activation,” PLoS One 17, no. 6 (2022): e0270008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Javaid N., Pham T. L. H., and Choi S., “Functional Comparison Between VP64‐dCas9‐VP64 and dCas9‐VP192 CRISPR Activators in Human Embryonic Kidney Cells,” International Journal of Molecular Sciences 22, no. 1 (2021): 397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Syding L. A., Nickl P., Kasparek P., and Sedlacek R., “CRISPR/Cas9 Epigenome Editing Potential for Rare Imprinting Diseases: A Review,” Cells 9, no. 4 (2020): 993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Ansari I., Chaturvedi A., Chitkara D., and Singh S., “CRISPR/Cas Mediated Epigenome Editing for Cancer Therapy,” Seminars in Cancer Biology 83 (2022): 570–583. [DOI] [PubMed] [Google Scholar]
- 47. Alkan F., Wenzel A., Anthon C., et al., “CRISPR‐Cas9 Off‐Targeting Assessment With Nucleic Acid Duplex Energy Parameters,” Genome Biology 19, no. 1 (2018): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Hao Y., “CRISPR‐Cas System: Off‐Target Effects and Its Possible Solutions,” (2024), Third International Conference on Biological Engineering and Medical Science (ICBioMed2023), SPIE.
- 49. Cai Y., Chen L., Sun S., et al., “CRISPR/Cas9‐Mediated Deletion of Large Genomic Fragments in Soybean,” International Journal of Molecular Sciences 19, no. 12 (2018): 3835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Guilinger J. P., Pattanayak V., Reyon D., et al., “Broad Specificity Profiling of TALENs Results in Engineered Nucleases With Improved DNA‐Cleavage Specificity,” Nature Methods 11, no. 4 (2014): 429–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Manghwar H., Li B., Ding X., et al., “CRISPR/Cas Systems in Genome Editing: Methodologies and Tools for sgRNA Design, Off‐Target Evaluation, and Strategies to Mitigate Off‐Target Effects,” Advanced Science 7, no. 6 (2020): 1902312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Naeem M., Majeed S., Hoque M. Z., and Ahmad I., “Latest Developed Strategies to Minimize the Off‐Target Effects in CRISPR‐Cas‐Mediated Genome Editing,” Cells 9, no. 7 (2020): 1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Atkins A., Chung C. H., Allen A. G., et al., “Off‐Target Analysis in Gene Editing and Applications for Clinical Translation of CRISPR/Cas9 in HIV‐1 Therapy,” Frontiers in Genome Editing 3 (2021): 673022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lei Z., Meng H., Zhuang Y., Zhu Q., and Yi C., “Chemical and Biological Approaches to Interrogate Off‐Target Effects of Genome Editing Tools,” ACS Chemical Biology 18, no. 2 (2023): 205–217. [DOI] [PubMed] [Google Scholar]
- 55. Lino C. A., Harper J. C., Carney J. P., and Timlin J. A., “Delivering CRISPR: A Review of the Challenges and Approaches,” Drug Delivery 25, no. 1 (2018): 1234–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Yip B. H., “Recent Advances in CRISPR/Cas9 Delivery Strategies,” Biomolecules 10, no. 6 (2020): 839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Dai W. J., Zhu L. Y., Yan Z. Y., Xu Y., Wang Q. L., and Lu X. J., “CRISPR‐Cas9 for In Vivo Gene Therapy: Promise and Hurdles,” Molecular Therapy ‐ Nucleic Acids 5, no. 8 (2016): e349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Xu Y. and Li Z., “CRISPR‐Cas Systems: Overview, Innovations and Applications in Human Disease Research and Gene Therapy,” Computational and Structural Biotechnology Journal 18 (2020): 2401–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Li Y., Glass Z., Huang M., Chen Z. Y., and Xu Q., “Ex Vivo Cell‐Based CRISPR/Cas9 Genome Editing for Therapeutic Applications,” Biomaterials 234 (2020): 119711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Staal F. J. T., Aiuti A., and Cavazzana M., “Autologous Stem‐Cell‐Based Gene Therapy for Inherited Disorders: State of the Art and Perspectives,” Frontiers in Pediatrics 7 (2019): 443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Davodabadi F., Farasati Far B., Sargazi S., et al., “Nanomaterials‐Based Targeting of Long Non‐Coding RNAs in Cancer: A Cutting‐Edge Review of Current Trends,” ChemMedChem 19, no. 8 (2024): e202300528. [DOI] [PubMed] [Google Scholar]
- 62. Ou X., Ma Q., Yin W., Ma X., and He Z., “CRISPR/Cas9 Gene‐Editing in Cancer Immunotherapy: Promoting the Present Revolution in Cancer Therapy and Exploring More,” Frontiers in Cell and Development Biology 9 (2021): 674467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Liu Z., Shi M., Ren Y., et al., “Recent Advances and Applications of CRISPR‐Cas9 in Cancer Immunotherapy,” Molecular Cancer 22, no. 1 (2023): 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Dong A. C. and Rivella S., “Gene Addition Strategies for Beta‐Thalassemia and Sickle Cell Anemia,” Advances in Experimental Medicine and Biology 1013 (2017): 155–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Harteveld C. L., Achour A., Arkesteijn S. J. G., et al., “The Hemoglobinopathies, Molecular Disease Mechanisms and Diagnostics,” International Journal of Laboratory Hematology 44, no. 1 (2022): 28–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Otroj M., Taymouri S., Varshosaz J., and Mirian M., “Preparation and Characterization of Dry Powder Containing Sunitinib Loaded PHBV Nanoparticles for Enhanced Pulmonary Delivery,” Journal of Drug Delivery Science and Technology 56 (2020): 101570. [Google Scholar]
- 67. Lin H., Li G., Peng X., et al., “The Use of CRISPR/Cas9 as a Tool to Study Human Infectious Viruses,” Frontiers in Cellular and Infection Microbiology 11 (2021): 590989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Baddeley H. J. E. and Isalan M., “The Application of CRISPR/Cas Systems for Antiviral Therapy,” Frontiers in Genome Editing 3 (2021): 745559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kotagama O. W., Jayasinghe C. D., and Abeysinghe T., “Era of Genomic Medicine: A Narrative Review on CRISPR Technology as a Potential Therapeutic Tool for Human Diseases,” BioMed Research International 2019 (2019): 1369682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. De Masi C., Spitalieri P., Murdocca M., et al., “Application of CRISPR/Cas9 to Human‐Induced Pluripotent Stem Cells: From Gene Editing to Drug Discovery,” Human Genomics 14, no. 1 (2020): 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Geng B. C., Choi K. H., Wang S. Z., et al., “A Simple, Quick, and Efficient CRISPR/Cas9 Genome Editing Method for Human Induced Pluripotent Stem Cells,” Acta Pharmacologica Sinica 41, no. 11 (2020): 1427–1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. McCloskey A. G., Miskelly M. G., Moore C. B. T., et al., “CRISPR/Cas9 Gene Editing Demonstrates Metabolic Importance of GPR55 in the Modulation of GIP Release and Pancreatic Beta Cell Function,” Peptides 125 (2020): 170251. [DOI] [PubMed] [Google Scholar]
- 73. Burnight E. R., Gupta M., Wiley L. A., et al., “Using CRISPR‐Cas9 to Generate Gene‐Corrected Autologous iPSCs for the Treatment of Inherited Retinal Degeneration,” Molecular Therapy 25, no. 9 (2017): 1999–2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Dominguez A. A., Lim W. A., and Qi L. S., “Beyond Editing: Repurposing CRISPR–Cas9 for Precision Genome Regulation and Interrogation,” Nature Reviews Molecular Cell Biology 17, no. 1 (2016): 5–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Xu X., Chemparathy A., Zeng L., et al., “Engineered Miniature CRISPR‐Cas System for Mammalian Genome Regulation and Editing,” Molecular Cell 81, no. 20 (2021): 4333–4345. [DOI] [PubMed] [Google Scholar]
- 76. Song P., Zhang Q., Xu Z., Shi Y., Jing R., and Luo D., “CRISPR/Cas‐Based CAR‐T Cells: Production and Application,” Biomarker Research 12, no. 1 (2024): 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Wei W., Chen Z.‐N., and Wang K., “CRISPR/Cas9: A Powerful Strategy to Improve CAR‐T Cell Persistence,” International Journal of Molecular Sciences 24, no. 15 (2023): 12317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Lei T., Wang Y., Zhang Y., et al., “Leveraging CRISPR Gene Editing Technology to Optimize the Efficacy, Safety and Accessibility of CAR T‐Cell Therapy,” Leukemia 38 (2024): 1–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Zhang B., “CRISPR/Cas Gene Therapy,” Journal of Cellular Physiology 236, no. 4 (2021): 2459–2481. [DOI] [PubMed] [Google Scholar]
- 80. Rogozin I. B., Pavlov Y. I., Goncearenco A., et al., “Mutational Signatures and Mutable Motifs in Cancer Genomes,” Briefings in Bioinformatics 19, no. 6 (2018): 1085–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Kiri S. and Ryba T., “Cancer, Metastasis, and the Epigenome,” Molecular Cancer 23, no. 1 (2024): 154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Raj‐Kumar P.‐K., Lin X., Liu T., et al., “Proteogenomic Characterization of Difficult‐To‐Treat Breast Cancer With Tumor Cells Enriched Through Laser Microdissection,” Breast Cancer Research 26, no. 1 (2024): 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Yin Y., Sapida J., Sukovich D., Patterson D., and Tentori A., “Unraveling Spatial Complexity of the Tumor Microenvironment: A Whole Transcriptomic Perspective With Visium HD,” Cancer Research 84, no. 6_Supplement (2024): 3645. [Google Scholar]
- 84. Goenka A., Khan F., Verma B., et al., “Tumor Microenvironment Signaling and Therapeutics in Cancer Progression,” Cancer Communications 43, no. 5 (2023): 525–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Rosenblum D., Gutkin A., Kedmi R., et al., “CRISPR‐Cas9 Genome Editing Using Targeted Lipid Nanoparticles for Cancer Therapy,” Science Advances 6 (2020): eabc9450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Chen Z., Liu F., Chen Y., et al., “Targeted Delivery of CRISPR/Cas9‐Mediated Cancer Gene Therapy via Liposome‐Templated Hydrogel Nanoparticles,” Advanced Functional Materials 27 (2017): 1703036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Zhang L., Kim D., and Oh Y. K., “Lipid Nanoparticle‐Mediated Efficient Delivery of CRISPR/Cas9 for Tumor Therapy,” NPG Asia Materials 17, no. 5 (2017): 641–652. [Google Scholar]
- 88. Behan F. M., Iorio F., Gonçalves E., et al., “Prioritisation of Oncology Therapeutic Targets Using CRISPR‐Cas9 Screening,” bioRxiv, 2018, 502005.
- 89. Meca‐Cortés O., Guerra‐Rebollo M., Garrido C., Borrós S., Rubio N., and Blanco J., “CRISPR/Cas9‐Mediated Knockin Application in Cell Therapy: A Non‐Viral Procedure for Bystander Treatment of Glioma in Mice,” Molecular Therapy—Nucleic Acids 8 (2017): 395–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Liang C., Li F., Wang L., et al., “Tumor Cell‐Targeted Delivery of CRISPR/Cas9 by Aptamer‐Functionalized Lipopolymer for Therapeutic Genome Editing of VEGFA in Osteosarcoma,” Biomaterials 147 (2017): 68–85. [DOI] [PubMed] [Google Scholar]
- 91. Wang G., Chow R. D., Bai Z., et al., “Multiplexed Activation of Endogenous Genes by CRISPRa Elicits Potent Anti‐Tumor Immunity,” Nature Immunology 20 (2019): 1494–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Tang H. and Shrager J. B., “CRISPR/Cas‐Mediated Genome Editing to Treat EGFR‐Mutant Lung Cancer: A Personalized Molecular Surgical Therapy,” EMBO Molecular Medicine 8 (2016): 83–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Zhang B., Wu P.‐Y., Zou J.‐J., et al., “Efficient CRISPR/Cas9 Gene‐Chemo Synergistic Cancer Therapy via a Stimuli‐Responsive Chitosan‐Based Nanocomplex Elicits Anti‐Tumorigenic Pathway Effect,” Chemical Engineering Journal 393 (2020): 124688. [Google Scholar]
- 94. Zhang L., Li Y., Chen Q., Xia Y., Zheng W., and Jiang X., “The Construction of Drug‐Resistant Cancer Cell Lines by CRISPR/Cas9 System for Drug Screening,” Science Bulletin 63, no. 21 (2018): 1411–1419. [DOI] [PubMed] [Google Scholar]
- 95. Rayner E., Durin M. A., Thomas R., et al., “CRISPR‐Cas9 Causes Chromosomal Instability and Rearrangements in Cancer Cell Lines, Detectable by Cytogenetic Methods,” CRISPR Journal 2 (2019): 406–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Kouranova E., Forbes K., Zhao G., et al., “vCRISPRs for Optimal Targeting: Delivery of CRISPR Components as DNA, RNA, and Protein Into Cultured Cells and Single‐Cell Embryos,” Human Gene Therapy 27 (2016): 464–475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Deng H., Tan S., Gao X., et al., “Cdk5 Knocking out Mediated by CRISPR‐Cas9 Genome Editing for PD‐L1 Attenuation and Enhanced Antitumor Immunity,” Acta Pharmaceutica Sinica B 10 (2019): 358–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Szlachta K., Kuscu C., Tufan T., et al., “CRISPR Knockout Screening Identifies Combinatorial Drug Targets in Pancreatic Cancer and Models Cellular Drug Response,” Nature Communications 9 (2018): 4275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Lin S., Larrue C., Scheidegger N. K., et al., “An In Vivo CRISPR Screening Platform for Prioritizing Therapeutic Targets in AML,” Cancer Discovery 12 (2020): 432–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Liu Y., Zhao G., Xu C. F., Luo Y. L., Lu Z. D., and Wang J., “Systemic Delivery of CRISPR/Cas9 With PEG‐PLGA Nanoparticles for Chronic Myeloid Leukemia Targeted Therapy,” Biomaterials Science 6, no. 6 (2018): 1592–1603. [DOI] [PubMed] [Google Scholar]
- 101. Cheung A. H.‐K., Chow C., Zhang J., et al., “Specific Targeting of Point Mutations in EGFR L858R‐Positive Lung Cancer by CRISPR/Cas9,” Laboratory Investigation 98 (2018): 968–976. [DOI] [PubMed] [Google Scholar]
- 102. Wang H. and Sun W., “CRISPR‐Mediated Targeting of HER2 Inhibits Cell Proliferation Through a Dominant Negative Mutation,” Cancer Letters 385 (2017): 137–143. [DOI] [PubMed] [Google Scholar]
- 103. Ibrahim A. U., “Genome Engineering Using the CRISPR Cas9 System,” 2019.
- 104. Cook P. J. and Ventura A., “Cancer Diagnosis and Immunotherapy in the Age of CRISPR,” Genes 58 (2019): 233–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. He S., “Development of Novel Inducible CRISPR Tools and the Application to Study DNA Damage Responses,” 2023. Johns Hopkins University.
- 106. Wang X., “Gene Mutation‐Based and Specific Therapies in Precision Medicine,” Journal of Cellular and Molecular Medicine 20 (2016): 577–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Noor A., Bilal A., and Ali U., “Towards Personalized Cancer Care: A Report of CRISPR‐Cas9 Applications in Targeted Therapies and Precision Medicine,” Journal of Health and Rehabilitation Research 4, no. 2 (2024): 1375–1380. [Google Scholar]
- 108. Sharma A. K. and Giri A. K., “Engineering CRISPR/Cas9 Therapeutics for Cancer Precision Medicine,” Frontiers in Genetics 15 (2024): 1309175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Lara‐Sáez I., Mencía Á., Recuero E., et al., “Nonviral CRISPR/Cas9 Mutagenesis for Streamlined Generation of Mouse Lung Cancer Models,” Proceedings of the National Academy of Sciences 121, no. 28 (2024): e2322917121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Park T., Ye S., Shin S. K., Kim K., Hur J. K., and Hur J. W., “CRISPR‐Cas9‐Generated Mouse Model of Neurofibromatosis Type 1,” Molecular & Cellular Toxicology 19, no. 2 (2023): 277–282. [Google Scholar]
- 111. Park H., Kang Y. K., and Shim G., “CRISPR/Cas9‐Mediated Customizing Strategies for Adoptive T‐Cell Therapy,” Pharmaceutics 16, no. 3 (2024): 346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Chen X., Zhong S., Zhan Y., and Zhang X., “CRISPR‐Cas9 Applications in T Cells and Adoptive T Cell Therapies,” Cellular & Molecular Biology Letters 29, no. 1 (2024): 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Stefanoudakis D., Kathuria‐Prakash N., Sun A. W., et al., “The Potential Revolution of Cancer Treatment With CRISPR Technology,” Cancers 15, no. 6 (2023): 1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Dey S., Devender M., Rani S., and Pandey R. K., “Recent Advances in CAR T‐Cell Engineering Using Synthetic Biology: Paving the Way for Next‐Generation Cancer Treatment,” Advances in Protein Chemistry and Structural Biology 140 (2024): 91–156. [DOI] [PubMed] [Google Scholar]
- 115. Uscanga‐Palomeque A. C., Chávez‐Escamilla A. K., Alvizo‐Báez C. A., et al., “CAR‐T Cell Therapy: From the Shop to Cancer Therapy,” International Journal of Molecular Sciences 24, no. 21 (2023): 15688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Depil S., Duchateau P., Grupp S. A., Mufti G., and Poirot L., “'Off‐The‐Shelf' Allogeneic CAR T Cells: Development and Challenges,” Nature Reviews. Drug Discovery 19, no. 3 (2020): 185–199. [DOI] [PubMed] [Google Scholar]
- 117. Mohty R. and Lazaryan A., “‘off‐The‐Shelf’ Allogeneic Chimeric Antigen Receptor T‐Cell Therapy for B‐Cell Malignancies: Current Clinical Evidence and Challenges,” Frontiers in Oncology 14 (2024): 1433432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Dimitri A., Herbst F., and Fraietta J. A., “Engineering the Next‐Generation of CAR T‐Cells With CRISPR‐Cas9 Gene Editing,” Molecular Cancer 21, no. 1 (2022): 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Ren J., Zhang X., Liu X., et al., “A Versatile System for Rapid Multiplex Genome‐Edited CAR T Cell Generation,” Oncotarget 8, no. 10 (2017): 17002–17011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Al Abbar A., Ngai S. C., Nograles N., et al., “Induced Pluripotent Stem Cells: Reprogramming Platforms and Applications in Cell Replacement Therapy,” Bioresearch Open Access 9, no. 1 (2020): 121–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Afolabi L. O., Adeshakin A. O., Sani M. M., Bi J., and Wan X., “Genetic Reprogramming for NK Cell Cancer Immunotherapy With CRISPR/Cas9,” Immunology 158, no. 2 (2019): 63–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Sekiba K., Yamagami M., Otsuka M., et al., “Transcriptional Activation of the MICA Gene With an Engineered CRISPR‐Cas9 System,” Biochemical and Biophysical Research Communications 486, no. 2 (2017): 521–525. [DOI] [PubMed] [Google Scholar]
- 123. Hu W., Zi Z., Jin Y., et al., “CRISPR/Cas9‐Mediated PD‐1 Disruption Enhances Human Mesothelin‐Targeted CAR T Cell Effector Functions,” Cancer Immunology, Immunotherapy 68, no. 3 (2019): 365–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Guo X., Jiang H., Shi B., et al., “Disruption of PD‐1 Enhanced the Anti‐Tumor Activity of Chimeric Antigen Receptor T Cells Against Hepatocellular Carcinoma,” Frontiers in Pharmacology 9 (2018): 1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Landrum M. J., Lee J. M., Benson M., et al., “ClinVar: Public Archive of Interpretations of Clinically Relevant Variants,” Nucleic Acids Research 44, no. D1 (2016): D862–D868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Hu W., Wang G., Huang D., et al., “Cancer Immunotherapy Based on Natural Killer Cells: Current Progress and New Opportunities,” Frontiers in Immunology 10 (2019): 1205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Li Y., Hermanson D. L., Moriarity B. S., and Kaufman D. S., “Human iPSC‐Derived Natural Killer Cells Engineered With Chimeric Antigen Receptors Enhance Anti‐Tumor Activity,” Cell Stem Cell 23, no. 2 (2018): 181–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. “Fate Therapeutics Announces First Patient Treated With IPSC‐Derived NK Cell Cancer Immunotherapy FT500 Successfully Completes Initial Safety Assessment,” (2019), https://pipelinereview.com/fate‐therapeutics‐announces‐first‐patient‐treated‐with‐ipsc‐derived‐nk‐cell‐cancer‐immunotherapy‐ft500‐successfully‐completes‐initial‐safety‐assessment/.
- 129. Lin X., Sun Y., Dong X., Liu Z., Sugimura R., and Xie G., “IPSC‐Derived CAR‐NK Cells for Cancer Immunotherapy,” Biomedicine & Pharmacotherapy 165 (2023): 115123. [DOI] [PubMed] [Google Scholar]
- 130. Yang C., Sun C., Tan B., et al., “Allogeneic Anti‐CD19 CAR‐T Cells Induce Remission in Refractory Systemic Lupus Erythematosus,” Cell Research 35 (2025): 1–3, 10.1038/s41422-025-01128-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Mougiakakos D., Krönke G., Völkl S., et al., “CD19‐Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus,” New England Journal of Medicine 385, no. 6 (2021): 567–569. [DOI] [PubMed] [Google Scholar]
- 132. Liu J., Zhao Y., and Zhao H., “Chimeric Antigen Receptor T‐Cell Therapy in Autoimmune Diseases,” Frontiers in Immunology 15 (2024): 1492552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Berry C. T., Frazee C. S., Herman P. J., et al., “Current Advancements in Cellular Immunotherapy for Autoimmune Disease,” Seminars in Immunopathology 47, no. 1 (2025): 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Zhang Y., Wei J., Cao J., et al., “Requirements for Human‐Induced Pluripotent Stem Cells,” Cell Proliferation 55, no. 4 (2022): e13182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Hao J., Ma A., Wang L., et al., “General Requirements for Stem Cells,” Cell Proliferation 53, no. 12 (2020): e12926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Frangoul H., Altshuler D., Cappellini M. D., et al., “CRISPR‐Cas9 Gene Editing for Sickle Cell Disease and β‐Thalassemia,” New England Journal of Medicine 384, no. 3 (2021): 252–260. [DOI] [PubMed] [Google Scholar]
- 137. Huang X., Wang Y., Yan W., et al., “Production of Gene‐Corrected Adult Beta Globin Protein in Human Erythrocytes Differentiated From Patient iPSCs After Genome Editing of the Sickle Point Mutation,” Stem Cells 33, no. 5 (2015): 1470–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Smith C., Abalde‐Atristain L., He C., et al., “Efficient and Allele‐Specific Genome Editing of Disease Loci in Human iPSCs,” Molecular Therapy 23, no. 3 (2015): 570–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Xie F., Ye L., Chang J. C., et al., “Seamless Gene Correction of β‐Thalassemia Mutations in Patient‐Specific iPSCs Using CRISPR/Cas9 and piggyBac,” Genome Research 24, no. 9 (2014): 1526–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Wu Y., Liang D., Wang Y., et al., “Correction of a Genetic Disease in Mouse via Use of CRISPR‐Cas9,” Cell Stem Cell 13, no. 6 (2013): 659–662. [DOI] [PubMed] [Google Scholar]
- 141. Osborn M. J., Gabriel R., Webber B. R., et al., “Fanconi Anemia Gene Editing by the CRISPR/Cas9 System,” Human Gene Therapy 26, no. 2 (2015): 114–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Pan Y., Shen N., Jung‐Klawitter S., et al., “CRISPR RNA‐Guided FokI Nucleases Repair a PAH Variant in a Phenylketonuria Model,” Scientific Reports 6 (2016): 35794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Firth A. L., Menon T., Parker G. S., et al., “Functional Gene Correction for Cystic Fibrosis in Lung Epithelial Cells Generated From Patient iPSCs,” Cell Reports 12, no. 9 (2015): 1385–1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Yang Y., Wang L., Bell P., et al., “A Dual AAV System Enables the Cas9‐Mediated Correction of a Metabolic Liver Disease in Newborn Mice,” Nature Biotechnology 34, no. 3 (2016): 334–338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Kang Y., Zheng B., Shen B., et al., “CRISPR/Cas9‐Mediated Dax1 Knockout in the Monkey Recapitulates Human AHC‐HH,” Human Molecular Genetics 24, no. 25 (2015): 7255–7264. [DOI] [PubMed] [Google Scholar]
- 146. Yin H., Xue W., Chen S., et al., “Genome Editing With Cas9 in Adult Mice Corrects a Disease Mutation and Phenotype,” Nature Biotechnology 32, no. 6 (2014): 551–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Feng Y., Sassi S., Shen J. K., et al., “Targeting CDK11 in Osteosarcoma Cells Using the CRISPR‐Cas9 System,” Journal of Orthopaedic Research 33, no. 2 (2015): 199–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Matano M., Date S., Shimokawa M., et al., “Modeling Colorectal Cancer Using CRISPR‐Cas9‐Mediated Engineering of Human Intestinal Organoids,” Nature Medicine 21, no. 3 (2015): 256–262. [DOI] [PubMed] [Google Scholar]
- 149. Platt R. J., Chen S., Zhou Y., et al., “CRISPR‐Cas9 Knockin Mice for Genome Editing and Cancer Modeling,” Cell 159, no. 2 (2014): 440–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Ding Q., Strong A., Patel K. M., et al., “Permanent Alteration of PCSK9 With In Vivo CRISPR‐Cas9 Genome Editing,” Circulation Research 115, no. 5 (2014): 488–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Yang L., Grishin D., Wang G., et al., “Targeted and Genome‐Wide Sequencing Reveal Single Nucleotide Variations Impacting Specificity of Cas9 in Human Stem Cells,” Nature Communications 5 (2014): 5507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Dow L. E., Fisher J., O'Rourke K. P., et al., “Inducible In Vivo Genome Editing With CRISPR‐Cas9,” Nature Biotechnology 33, no. 4 (2015): 390–394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Carroll K. J., Makarewich C. A., McAnally J., et al., “A Mouse Model for Adult Cardiac‐Specific Gene Deletion With CRISPR/Cas9,” Proceedings of the National Academy of Sciences of the United States of America 113, no. 2 (2016): 338–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Bloh K. M., Bialk P. A., Gopalakrishnapillai A., Kolb E. A., and Kmiec E. B., “CRISPR/Cas9‐Directed Reassignment of the GATA1 Initiation Codon in K562 Cells to Recapitulate AML in Down Syndrome,” Molecular Therapy–Nucleic Acids 7 (2017): 288–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Bassuk A. G., Zheng A., Li Y., Tsang S. H., and Mahajan V. B., “Precision Medicine: Genetic Repair of Retinitis Pigmentosa in Patient‐Derived Stem Cells,” Scientific Reports 6 (2016): 19969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Swiech L., Heidenreich M., Banerjee A., et al., “In Vivo Interrogation of Gene Function in the Mammalian Brain Using CRISPR‐Cas9,” Nature Biotechnology 33, no. 1 (2015): 102–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Long C., McAnally J. R., Shelton J. M., Mireault A. A., Bassel‐Duby R., and Olson E. N., “Prevention of Muscular Dystrophy in Mice by CRISPR/Cas9‐Mediated Editing of Germline DNA,” Science 345, no. 6201 (2014): 1184–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Xu L., Park K. H., Zhao L., et al., “CRISPR‐Mediated Genome Editing Restores Dystrophin Expression and Function in Mdx Mice,” Molecular Therapy 24, no. 3 (2016): 564–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. El Refaey M., Xu L., Gao Y., et al., “In Vivo Genome Editing Restores Dystrophin Expression and Cardiac Function in Dystrophic Mice,” Circulation Research 121, no. 8 (2017): 923–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Petkova M. V., Stantzou A., Morin A., et al., “Live‐Imaging of Revertant and Therapeutically Restored Dystrophin in the Dmd(EGFP‐Mdx) Mouse Model for Duchenne Muscular Dystrophy,” Neuropathology and Applied Neurobiology 46, no. 6 (2020): 602–614. [DOI] [PubMed] [Google Scholar]
- 161. Lee K., Conboy M., Park H. M., et al., “Nanoparticle Delivery of Cas9 Ribonucleoprotein and Donor DNA In Vivo Induces Homology‐Directed DNA Repair,” Nature Biomedical Engineering 1 (2017): 889–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Ousterout D. G., Kabadi A. M., Thakore P. I., Majoros W. H., Reddy T. E., and Gersbach C. A., “Multiplex CRISPR/Cas9‐Based Genome Editing for Correction of Dystrophin Mutations That Cause Duchenne Muscular Dystrophy,” Nature Communications 6 (2015): 6244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Wang L., Yi F., Fu L., et al., “CRISPR/Cas9‐Mediated Targeted Gene Correction in Amyotrophic Lateral Sclerosis Patient iPSCs,” Protein & Cell 8, no. 5 (2017): 365–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Wang H., Guo W., Mitra J., et al., “Mutant FUS Causes DNA Ligation Defects to Inhibit Oxidative Damage Repair in Amyotrophic Lateral Sclerosis,” Nature Communications 9, no. 1 (2018): 3683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Zhang Y., Schmid B., Nikolaisen N. K., et al., “Patient iPSC‐Derived Neurons for Disease Modeling of Frontotemporal Dementia With Mutation in CHMP2B,” Stem Cell Reports 8, no. 3 (2017): 648–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Lopez‐Gonzalez R., Yang D., Pribadi M., et al., “Partial Inhibition of the Overactivated Ku80‐Dependent DNA Repair Pathway Rescues Neurodegeneration in C9ORF72‐ALS/FTD,” Proceedings of the National Academy of Sciences of the United States of America 116, no. 19 (2019): 9628–9633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Krishnan G., Zhang Y., Gu Y., Kankel M. W., Gao F. B., and Almeida S., “CRISPR Deletion of the C9ORF72 Promoter in ALS/FTD Patient Motor Neurons Abolishes Production of Dipeptide Repeat Proteins and Rescues Neurodegeneration,” Acta Neuropathologica 140, no. 1 (2020): 81–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Ababneh N. A., Scaber J., Flynn R., et al., “Correction of Amyotrophic Lateral Sclerosis Related Phenotypes in Induced Pluripotent Stem Cell‐Derived Motor Neurons Carrying a Hexanucleotide Expansion Mutation in C9orf72 by CRISPR/Cas9 Genome Editing Using Homology‐Directed Repair,” Human Molecular Genetics 29, no. 13 (2020): 2200–2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Colasante G., Lignani G., Brusco S., et al., “dCas9‐Based Scn1a Gene Activation Restores Inhibitory Interneuron Excitability and Attenuates Seizures in Dravet Syndrome Mice,” Molecular Therapy 28, no. 1 (2020): 235–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Hendel A., Bak R. O., Clark J. T., et al., “Chemically Modified Guide RNAs Enhance CRISPR‐Cas Genome Editing in Human Primary Cells,” Nature Biotechnology 33, no. 9 (2015): 985–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Yuan J., Ma Y., Huang T., et al., “Genetic Modulation of RNA Splicing With a CRISPR‐Guided Cytidine Deaminase,” Molecular Cell 72, no. 2 (2018): 380–394. [DOI] [PubMed] [Google Scholar]
- 172. Abudayyeh O. O., Gootenberg J. S., Essletzbichler P., et al., “RNA Targeting With CRISPR‐Cas13,” Nature 550, no. 7675 (2017): 280–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Oakes B. L., Fellmann C., Rishi H., et al., “CRISPR‐Cas9 Circular Permutants as Programmable Scaffolds for Genome Modification,” Cell 176, no. 1–2 (2019): 254–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Xie N., Zhou Y., Sun Q., et al., “Novel Epigenetic Techniques Provided by the CRISPR/Cas9 System,” Stem Cells International 2018 (2018): 7834175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Yang L., Güell M., Niu D., et al., “Genome‐Wide Inactivation of Porcine Endogenous Retroviruses (PERVs),” Science 350, no. 6264 (2015): 1101–1104. [DOI] [PubMed] [Google Scholar]
- 176. Chen J. S., Ma E., Harrington L. B., et al., “CRISPR‐Cas12a Target Binding Unleashes Indiscriminate Single‐Stranded DNase Activity,” Science 360, no. 6387 (2018): 436–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Niu D., Wei H. J., Lin L., et al., “Inactivation of Porcine Endogenous Retrovirus in Pigs Using CRISPR‐Cas9,” Science 357, no. 6357 (2017): 1303–1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Gootenberg J. S., Abudayyeh O. O., Lee J. W., et al., “Nucleic Acid Detection With CRISPR‐Cas13a/C2c2,” Science 356, no. 6336 (2017): 438–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Silva S., Pardee K., and Pena L., “Loop‐Mediated Isothermal Amplification (LAMP) for the Diagnosis of Zika Virus: A Review ,” Viruses 12, no. 1 (2019): 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Patchsung M., Jantarug K., Pattama A., et al., “Clinical Validation of a Cas13‐Based Assay for the Detection of SARS‐CoV‐2 RNA,” Nature Biomedical Engineering 4, no. 12 (2020): 1140–1149. [DOI] [PubMed] [Google Scholar]
- 181. Ding R., Long J., Yuan M., et al., “CRISPR/Cas System: A Potential Technology for the Prevention and Control of COVID‐19 and Emerging Infectious Diseases,” Frontiers in Cellular and Infection Microbiology 11 (2021): 639108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Garg N., Ahmad F. J., and Kar S., “Recent Advances in Loop‐Mediated Isothermal Amplification (LAMP) for Rapid and Efficient Detection of Pathogens,” Current Research in Microbial Sciences 3 (2022): 100120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Lin H., Li G., Peng X., et al., “The Use of CRISPR/Cas9 as a Tool to Study Human Infectious Viruses. Frontiers in Cellular and Infection,” Microbiology 11 (2021): 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Etienne L., Hahn B. H., Sharp P. M., Matsen F. A., and Emerman M., “Gene Loss and Adaptation to Hominids Underlie the Ancient Origin of HIV‐1,” Cell Host & Microbe 14, no. 1 (2013): 85–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Etienne L., Bibollet‐Ruche F., Sudmant P. H., Wu L. I., Hahn B. H., and Emerman M., “The Role of the Antiviral APOBEC3 Gene Family in Protecting Chimpanzees Against Lentiviruses From Monkeys,” PLoS Pathogens 11, no. 9 (2015): e1005149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Gélinas J.‐F., Gill D. R., and Hyde S. C., “Multiple Inhibitory Factors Act in the Late Phase of HIV‐1 Replication: A Systematic Review of the Literature,” Microbiology and Molecular Biology Reviews 82, no. 1 (2018): e00051‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Duggal N. K. and Emerman M., “Evolutionary Conflicts Between Viruses and Restriction Factors Shape Immunity,” Nature Reviews Immunology 12, no. 10 (2012): 687–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Arias J. F., Iwabu Y., and Tokunaga K., “Structural Basis for the Antiviral Activity of BST‐2/Tetherin and Its Viral Antagonism,” Frontiers in Microbiology 2 (2011): 250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Konermann S., Brigham M. D., Trevino A. E., et al., “Genome‐Scale Transcriptional Activation by an Engineered CRISPR‐Cas9 Complex,” Nature 517, no. 7536 (2015): 583–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zhang Y., Ozono S., Yao W., et al., “CRISPR‐Mediated Activation of Endogenous BST‐2/Tetherin Expression Inhibits Wild‐Type HIV‐1 Production,” Scientific Reports 9, no. 1 (2019): 3134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Noroozi Z., Shamsara M., Valipour E., et al., “Antiproliferative Effects of AAV‐Delivered CRISPR/Cas9‐Based Degradation of the HPV18‐E6 Gene in HeLa Cells,” Scientific Reports 12, no. 1 (2022): 2224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Gao C., Wu P., Yu L., et al., “The Application of CRISPR/Cas9 System in Cervical Carcinogenesis,” Cancer Gene Therapy 29, no. 5 (2022): 466–474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Teng M., Yao Y., Nair V., and Luo J., “Latest Advances of Virology Research Using CRISPR/Cas9‐Based Gene‐Editing Technology and Its Application to Vaccine Development,” Viruses 13, no. 5 (2021): 779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Shademan B., Nourazarian A., Hajazimian S., et al., “CRISPR Technology in Gene‐Editing‐Based Detection and Treatment of SARS‐CoV‐2,” Frontiers in Molecular Biosciences 8 (2021): 772788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Abbott T. R., Dhamdhere G., Liu Y., et al., “Development of CRISPR as an Antiviral Strategy to Combat SARS‐CoV‐2 and Influenza,” Cell 181, no. 4 (2020): 865–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Ye L., Wang J., Beyer A. I., et al., “Seamless Modification of Wild‐Type Induced Pluripotent Stem Cells to the Natural CCR5Δ32 Mutation Confers Resistance to HIV Infection,” Proceedings of the National Academy of Sciences of the United States of America 111, no. 26 (2014): 9591–9596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Ebina H., Misawa N., Kanemura Y., and Koyanagi Y., “Harnessing the CRISPR/Cas9 System to Disrupt Latent HIV‐1 Provirus,” Scientific Reports 3 (2013): 2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Wang J. and Quake S. R., “RNA‐Guided Endonuclease Provides a Therapeutic Strategy to Cure Latent Herpesviridae Infection,” Proceedings of the National Academy of Sciences of the United States of America 111, no. 36 (2014): 13157–13162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Hu W., Kaminski R., Yang F., et al., “RNA‐Directed Gene Editing Specifically Eradicates Latent and Prevents New HIV‐1 Infection,” Proceedings of the National Academy of Sciences of the United States of America 111, no. 31 (2014): 11461–11466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Lin S. R., Yang H. C., Kuo Y. T., et al., “The CRISPR/Cas9 System Facilitates Clearance of the Intrahepatic HBV Templates In Vivo,” Molecular Therapy ‐ Nucleic Acids 3, no. 8 (2014): e186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Ramanan V., Shlomai A., Cox D. B. T., et al., “CRISPR/Cas9 Cleavage of Viral DNA Efficiently Suppresses Hepatitis B Virus,” Scientific Reports 5 (2015): 10833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Vinayak S., Pawlowic M. C., Sateriale A., et al., “Genetic Modification of the Diarrhoeal Pathogen Cryptosporidium parvum ,” Nature 523, no. 7561 (2015): 477–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. van Diemen F. R., Kruse E. M., Hooykaas M. J. G., et al., “CRISPR/Cas9‐Mediated Genome Editing of Herpesviruses Limits Productive and Latent Infections,” PLoS Pathogens 12, no. 6 (2016): e1005701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Aimola G., Wight D. J., Flamand L., and Kaufer B. B., “Excision of Integrated Human Herpesvirus 6A Genomes Using CRISPR/Cas9 Technology,” Microbiology Spectrum 11, no. 2 (2023): e0076423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Xiao J., Deng J., Zhang Q., et al., “Targeting Human Cytomegalovirus IE Genes by CRISPR/Cas9 Nuclease Effectively Inhibits Viral Replication and Reactivation,” Archives of Virology 165, no. 8 (2020): 1827–1835. [DOI] [PubMed] [Google Scholar]
- 206. Wollebo H. S., Bellizzi A., Kaminski R., Hu W., White M. K., and Khalili K., “CRISPR/Cas9 System as an Agent for Eliminating Polyomavirus JC Infection,” PLoS One 10, no. 9 (2015): e0136046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Binnie A., Fernandes E., Almeida‐Lousada H., de Mello R. A., and Castelo‐Branco P., “CRISPR‐Based Strategies in Infectious Disease Diagnosis and Therapy,” Infection 49, no. 3 (2021): 377–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Kirby E. N., Shue B., Thomas P. Q., and Beard M. R., “CRISPR Tackles Emerging Viral Pathogens,” Viruses 13 (2021): 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Junaid M., Thirapanmethee K., Khuntayaporn P., and Chomnawang M. T., “CRISPR‐Based Gene Editing in Acinetobacter baumannii to Combat Antimicrobial Resistance,” Pharmaceuticals 16, no. 7 (2023): 920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Lee D., Muir P., Lundberg S., Lundholm A., Sandegren L., and Koskiniemi S., “A CRISPR‐Cas9 System Protecting E. coli Against Acquisition of Antibiotic Resistance Genes,” Scientific Reports 15, no. 1 (2025): 1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Mikkelsen K., Bowring J. Z., NG Y. K., et al., “An Endogenous Staphylococcus aureus CRISPR‐Cas System Limits Phage Proliferation and Is Efficiently Excised From the Genome as Part of the SCC Mec Cassette,” Microbiology Spectrum 11, no. 4 (2023): e01277–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Ciurea A. V., Mohan A. G., Covache‐Busuioc R. A., et al., “Unraveling Molecular and Genetic Insights Into Neurodegenerative Diseases: Advances in Understanding Alzheimer's, Parkinson's, and Huntington's Diseases and Amyotrophic Lateral Sclerosis,” International Journal of Molecular Sciences 24, no. 13 (2023): 10809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Kania K., Ambrosius W., Kozubski W., and Kalinowska‐Łyszczarz A., “The Impact of Disease Modifying Therapies on Cognitive Functions Typically Impaired in Multiple Sclerosis Patients: A Clinician's Review,” Frontiers in Neurology 14 (2023): 1222574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Datta A., Sarmah D., Kaur H., et al., “Advancement in CRISPR/Cas9 Technology to Better Understand and Treat Neurological Disorders,” Cellular and Molecular Neurobiology 43, no. 3 (2023): 1019–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. György B., Lööv C., Zaborowski M. P., et al., “CRISPR/Cas9 Mediated Disruption of the Swedish APP Allele as a Therapeutic Approach for Early‐Onset Alzheimer's Disease,” Molecular Therapy ‐ Nucleic Acids 11 (2018): 429–440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Ortiz‐Virumbrales M., Moreno C. L., Kruglikov I., et al., “CRISPR/Cas9‐Correctable Mutation‐Related Molecular and Physiological Phenotypes in iPSC‐Derived Alzheimer's PSEN2 (N141I) Neurons,” Acta Neuropathologica Communications 5, no. 1 (2017): 77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Raikwar S. P., Thangavel R., Dubova I., et al., “Targeted Gene Editing of Glia Maturation Factor in Microglia: A Novel Alzheimer's Disease Therapeutic Target,” Molecular Neurobiology 56, no. 1 (2019): 378–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Colasante G., Qiu Y., Massimino L., et al., “In Vivo CRISPRa Decreases Seizures and Rescues Cognitive Deficits in a Rodent Model of Epilepsy,” Brain 143, no. 3 (2020): 891–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Shin J. W., Kim K. H., Chao M. J., et al., “Permanent Inactivation of Huntington's Disease Mutation by Personalized Allele‐Specific CRISPR/Cas9,” Human Molecular Genetics 25, no. 20 (2016): 4566–4576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Kolli N., Lu M., Maiti P., Rossignol J., and Dunbar G., “CRISPR‐Cas9 Mediated Gene‐Silencing of the Mutant Huntingtin Gene in an In Vitro Model of Huntington's Disease,” International Journal of Molecular Sciences 18, no. 4 (2017): 754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Monteys A. M., Ebanks S. A., Keiser M. S., and Davidson B. L., “CRISPR/Cas9 Editing of the Mutant Huntingtin Allele In Vitro and In Vivo,” Molecular Therapy 25, no. 1 (2017): 12–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Yang S., Chang R., Yang H., et al., “CRISPR/Cas9‐Mediated Gene Editing Ameliorates Neurotoxicity in Mouse Model of Huntington's Disease,” Journal of Clinical Investigation 127, no. 7 (2017): 2719–2724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Kantor B., Tagliafierro L., Gu J., et al., “Downregulation of SNCA Expression by Targeted Editing of DNA Methylation: A Potential Strategy for Precision Therapy in PD,” Molecular Therapy 26, no. 11 (2018): 2638–2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Chen Y., Xiong M., Dong Y., et al., “Chemical Control of Grafted Human PSC‐Derived Neurons in a Mouse Model of Parkinson's Disease,” Cell Stem Cell 18, no. 6 (2016): 817–826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Lee J., Bayarsaikhan D., Arivazhagan R., et al., “CRISPR/Cas9 Edited sRAGE‐MSCs Protect Neuronal Death in Parkinson's Disease Model,” International Journal of Stem Cells 12, no. 1 (2019): 114–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Yao M., Yang Q., Lian M., et al., “Generation of Dip2a Homozygous Knockout Murine ES Cell Line IBMSe001‐A‐1 via CRISPR/Cas9 Technology,” Stem Cell Research 45 (2020): 101778. [DOI] [PubMed] [Google Scholar]
- 227. Saad F. A., Saad J. F., Siciliano G., Merlini L., and Angelini C., “Duchenne Muscular Dystrophy Gene Therapy,” Current Gene Therapy 24, no. 1 (2024): 17–28. [DOI] [PubMed] [Google Scholar]
- 228. Gruenewald T., Seeman T. E., Choo T. H., et al., “Cardiovascular Variability, Sociodemographics, and Biomarkers of Disease: The MIDUS Study,” Frontiers in Physiology 14 (2023): 1234427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Goell J. H. and Hilton I. B., “CRISPR/Cas‐Based Epigenome Editing: Advances, Applications, and Clinical Utility,” Trends in Biotechnology 39, no. 7 (2021): 678–691. [DOI] [PubMed] [Google Scholar]
- 230. Taniguchi J., Feng Y., Pandian G. N., et al., “Biomimetic Artificial Epigenetic Code for Targeted Acetylation of Histones,” Journal of the American Chemical Society 140, no. 23 (2018): 7108–7115. [DOI] [PubMed] [Google Scholar]
- 231. Gemberling M. P., Siklenka K., Rodriguez E., et al., “Transgenic Mice for In Vivo Epigenome Editing With CRISPR‐Based Systems,” Nature Methods 18, no. 8 (2021): 965–974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Mészár Z., Kókai É., Varga R., et al., “CRISPR/Cas9‐Based Mutagenesis of Histone H3. 1 in Spinal Dynorphinergic Neurons Attenuates Thermal Sensitivity in Mice,” International Journal of Molecular Sciences 23, no. 6 (2022): 3178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Singh A., Irfan H., Fatima E., Nazir Z., Verma A., and Akilimali A., “Revolutionary Breakthrough: FDA Approves CASGEVY, the First CRISPR/Cas9 Gene Therapy for Sickle Cell Disease,” Annals of Medicine and Surgery 86, no. 8 (2024): 4555–4559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Parums D. V., “First Regulatory Approvals for CRISPR‐Cas9 Therapeutic Gene Editing for Sickle Cell Disease and Transfusion‐Dependent β‐Thalassemia,” Medical Science Monitor: International Medical Journal of Experimental and Clinical Research 30 (2024): e944204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Kotit S., “Lessons From the First‐In‐Human In Vivo CRISPR/Cas9 Editing of the TTR Gene by NTLA‐2001 Trial in Patients With Transthyretin Amyloidosis With Cardiomyopathy,” Global Cardiology Science & Practice 2023, no. 1 (2023): e202304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Longhurst H. J., Lindsay K., Petersen R. S., et al., “CRISPR‐Cas9 In Vivo Gene Editing of KLKB1 for Hereditary Angioedema,” New England Journal of Medicine 390, no. 5 (2024): 432–441. [DOI] [PubMed] [Google Scholar]
- 237. O'Brien S., Nastoupil L. J., Essell J., et al., “A First‐In‐Human Phase 1, Multicenter, Open‐Label Study of CB‐010, a Next‐Generation CRISPR‐Edited Allogeneic Anti‐CD19 CAR‐T Cell Therapy With a PD‐1 Knockout, in Patients With Relapsed/Refractory b Cell Non‐Hodgkin Lymphoma (ANTLER Study),” Blood 140, no. Supplement 1 (2022): 9457–9458. [Google Scholar]
- 238. Pal S. K., Tran B., Haanen J. B. A. G., et al., “CD70‐Targeted Allogeneic CAR T‐Cell Therapy for Advanced Clear Cell Renal Cell Carcinoma,” Cancer Discovery 14, no. 7 (2024): 1176–1189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Horie T. and Ono K., “VERVE‐101: A promising CRISPR‐Based Gene Editing Therapy that Reduces LDL‐C and PCSK9 Levels in HeFH Patients,” European Heart Journal ‐ Cardiovascular Pharmacotherapy 10, no. 2 (2024): 89–90. [DOI] [PubMed] [Google Scholar]
- 240. “CRISPR Gene Therapy EBT‐101 Does Not Prevent HIV Viral Rebound,” (2024), https://www.aidsmap.com/news/may‐2024/crispr‐gene‐therapy‐ebt‐101‐does‐not‐prevent‐hiv‐viral‐rebound.
- 241. Khambhati K., Bhattacharjee G., and Singh V., “Current Progress in CRISPR‐Based Diagnostic Platforms,” Journal of Cellular Biochemistry 120, no. 3 (2019): 2721–2725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Li Y., Li S., Wang J., and Liu G., “CRISPR/Cas Systems Towards Next‐Generation Biosensing,” Trends in Biotechnology 37, no. 7 (2019): 730–743. [DOI] [PubMed] [Google Scholar]
- 243. Wang W., Hou J., Zheng N., et al., “Keeping Our Eyes on CRISPR: The ‘Atlas’ of Gene Editing,” Cell Biology and Toxicology 35 (2019): 285–288. [DOI] [PubMed] [Google Scholar]
- 244. Qiu X.‐Y., Zhu L. Y., Zhu C. S., et al., “Highly Effective and Low‐Cost microRNA Detection With CRISPR‐Cas9,” ACS Synthetic Biology 7, no. 3 (2018): 807–813. [DOI] [PubMed] [Google Scholar]
- 245. Li L., Shen G., Wu M., Jiang J., Xia Q., and Lin P., “CRISPR‐Cas‐Mediated Diagnostics,” Trends in Biotechnology 40 (2022): 1326–1345. [DOI] [PubMed] [Google Scholar]
- 246. Durán‐Vinet B., Araya‐Castro K., Calderón J., et al., “CRISPR/Cas13‐Based Platforms for a Potential Next‐Generation Diagnosis of Colorectal Cancer Through Exosomes Micro‐RNA Detection: A Review,” Cancers (Basel) 13, no. 18 (2021): 4640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Yang H., Bailey P., and Pilarsky C., “CRISPR Cas9 in Pancreatic Cancer Research,” Frontiers in Cell and Development Biology 7 (2019): 239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Ramaker R. C., Hardigan A. A., Gordon E. R., Wright C. A., Myers R. M., and Cooper S. J., “Pooled CRISPR Screening in Pancreatic Cancer Cells Implicates Co‐Repressor Complexes as a Cause of Multiple Drug Resistance via Regulation of Epithelial‐To‐Mesenchymal Transition,” BMC Cancer 21, no. 1 (2021): 632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Wei X., Yang J., Adair S. J., et al., “Targeted CRISPR Screening Identifies PRMT5 as Synthetic Lethality Combinatorial Target With Gemcitabine in Pancreatic Cancer Cells,” Proceedings of the National Academy of Sciences of the United States of America 117, no. 45 (2020): 28068–28079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Skripova V., Serebriiskii I., Kiyamova R., and Astsaturov I., “Application of CRISPR/Cas9 System for Identification of Genes Involved in the Regulation of Pancreatic Cancer Cells Platinum Sensitivity,” Annals of Oncology 29 (2018): viii46–viii47. [Google Scholar]
- 251. Skripova V., Vlasenkova R., Zhou Y., et al., “Identification of New Regulators of Pancreatic Cancer Cell Sensitivity to Oxaliplatin and Cisplatin,” Molecules 27, no. 4 (2022): 1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Hou J., He Z., Liu T., et al., “Evolution of Molecular Targeted Cancer Therapy: Mechanisms of Drug Resistance and Novel Opportunities Identified by CRISPR‐Cas9 Screening,” Frontiers in Oncology 12 (2022): 755053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Zhang F., Fan Q., Ren K., Auerbach A. D., and Andreassen P. R., “FANCJ/BRIP1 Recruitment and Regulation of FANCD2 in DNA Damage Responses,” Chromosoma 119, no. 6 (2010): 637–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Kim W., Lee S., Kim H. S., et al., “Targeting Mutant KRAS With CRISPR‐Cas9 Controls Tumor Growth,” Genome Research 28, no. 3 (2018): 374–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Bai L., Pang Y., Wang T., et al., “SPEAR: CRISPR‐Mediated Ultrasensitive, Specific and Rapid One‐Pot Detection Strategy for Cancer‐Related SNPs,” Theranostics 15, no. 8 (2025): 3275–3288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Lo J. H., Hao L., Muzumdar M. D., et al., “iRGD‐Guided Tumor‐Penetrating Nanocomplexes for Therapeutic siRNA Delivery to Pancreatic Cancer,” Molecular Cancer Therapeutics 17, no. 11 (2018): 2377–2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Nikitovic D., Kukovyakina E., Berdiaki A., et al., “Enhancing Tumor Targeted Therapy: The Role of iRGD Peptide in Advanced Drug Delivery Systems,” Cancers (Basel) 16, no. 22 (2024): 3768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Uppada V., Gokara M., and Rasineni G. K., “Diagnosis and Therapy With CRISPR Advanced CRISPR Based Tools for Point of Care Diagnostics and Early Therapies,” Gene 656 (2018): 22–29. [DOI] [PubMed] [Google Scholar]
- 259. Foss D. V., Hochstrasser M. L., and Wilson R. C., “Clinical Applications of CRISPR‐Based Genome Editing and Diagnostics,” Transfusion 59, no. 4 (2019): 1389–1399. [DOI] [PubMed] [Google Scholar]
- 260. Woloshin S., Patel N., and Kesselheim A. S., “False Negative Tests for SARS‐CoV‐2 Infection—Challenges and Implications,” New England Journal of Medicine 383, no. 6 (2020): e38. [DOI] [PubMed] [Google Scholar]
- 261. Palaz F., Kalkan A. K., Tozluyurt A., and Ozsoz M., “CRISPR‐Based Tools: Alternative Methods for the Diagnosis of COVID‐19,” Clinical Biochemistry 89 (2021): 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Eid A. and Mahfouz M. M., “Genome Editing: The Road of CRISPR/Cas9 From Bench to Clinic,” Experimental & Molecular Medicine 48, no. 10 (2016): e265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Hanna E., Rémuzat C., Auquier P., and Toumi M., “Gene Therapies Development: Slow Progress and Promising Prospect,” Journal of Market Access & Health Policy 5, no. 1 (2017): 1265293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Javaid N. and Choi S., “CRISPR/Cas System and Factors Affecting Its Precision and Efficiency,” Frontiers in Cell and Development Biology 9 (2021): 761709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Bashor C. J., Hilton I. B., Bandukwala H., Smith D. M., and Veiseh O., “Engineering the Next Generation of Cell‐Based Therapeutics,” Nature Reviews Drug Discovery 21, no. 9 (2022): 655–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Wangler M. F., Yamamoto S., Chao H. T., et al., “Model Organisms Facilitate Rare Disease Diagnosis and Therapeutic Research,” Genetics 207, no. 1 (2017): 9–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Szatanek R., Baj‐Krzyworzeka M., Zimoch J., Lekka M., Siedlar M., and Baran J., “The Methods of Choice for Extracellular Vesicles (EVs) Characterization,” International Journal of Molecular Sciences 18, no. 6 (2017): 1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. El Andaloussi S., Mäger I., Breakefield X. O., and Wood M. J. A., “Extracellular Vesicles: Biology and Emerging Therapeutic Opportunities,” Nature Reviews Drug Discovery 12, no. 5 (2013): 347–357. [DOI] [PubMed] [Google Scholar]
- 269. Colombo M., Raposo G., and Théry C., “Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles,” Annual Review of Cell and Developmental Biology 30 (2014): 255–289. [DOI] [PubMed] [Google Scholar]
- 270. Haney M. J., Klyachko N. L., Zhao Y., et al., “Exosomes as Drug Delivery Vehicles for Parkinson's Disease Therapy,” Journal of Controlled Release 207 (2015): 18–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Shen B., Zhang J., Wu H., et al., “Generation of Gene‐Modified Mice via Cas9/RNA‐Mediated Gene Targeting,” Cell Research 23, no. 5 (2013): 720–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1. Overview of viral and non‐viral CRISPR delivery methods and their characteristics. This table summarises a broad range of non‐viral delivery systems developed for transporting CRISPR components—including DNA, RNA, and ribonucleoprotein (RNP) complexes—into target cells. The methods are categorised by their underlying mechanisms (physical or chemical), and each entry details the delivery approach, cargo type, major advantages, and associated drawbacks. Technologies include well‐established platforms like electroporation and lipofection, as well as emerging nanotechnologies such as gold nanoparticles, DNA nanoclews, and multifunctional envelope‐type nano devices (MENDs). The table emphasises each method’s utility for in vitro or in vivo applications, editing efficiency, cytotoxicity, delivery specificity, and translational feasibility. This comparative overview supports the selection of optimal non‐viral delivery strategies in CRISPR‐based research and therapeutic development.
Table S2. Ongoing clinical trials and applications of CRISPR. Summary of significant clinical developments and recent studies using CRISPR‐based therapies. As of early 2025, this table lists notable instances of CRISPR technology applications through clinical trials. In a variety of disease areas, such as genetic disorders, oncology, cardiovascular disease, and infectious disease, it highlights a range of therapeutic approaches, such as ex vivo cell therapies, in vivo gene editing via nucleases base editors, and engineered cell therapies. Key reported discoveries or outcomes, the target indication, the current clinical trial phase, the particular therapy or application, and pertinent citations are all included in the details.
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.
