Next Article in Journal
The Oral Microbiota: Implications in Mucosal Health and Systemic Disease—Crosstalk with Gut and Brain
Previous Article in Journal
Ovulation-Derived Fibronectin Promotes Peritoneal Seeding of High-Grade Serous Carcinoma Precursor Cells via Integrin β1 Signaling
Previous Article in Special Issue
Advances in β-Thalassemia Gene Therapy: CRISPR/Cas Systems and Delivery Innovations
error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Genome Safe Harbor Sites: A Comprehensive Review of Criteria, Discovery, Features, and Applications

1
Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
2
Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy
3
Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
4
Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH 43210, USA
*
Authors to whom correspondence should be addressed.
Cells 2026, 15(1), 81; https://doi.org/10.3390/cells15010081
Submission received: 4 December 2025 / Revised: 26 December 2025 / Accepted: 31 December 2025 / Published: 4 January 2026
(This article belongs to the Special Issue CRISPR-Based Genome Editing in Translational Research—Third Edition)

Abstract

The stable and safe integration of exogenous DNA into the genome is crucial to both genetic engineering and gene therapy. Traditional transgenesis approaches, such as those using retroviral vectors, result in random genomic integration, posing the risk of insertional mutagenesis and transcriptional dysregulation. Safe harbor sites (SHSs), genomic loci that support reliable transgene expression without compromising endogenous gene function, genomic integrity, or cellular physiology, have been identified and characterized across various model organisms. Well-established SHSs such as AAVS1, ROSA26, and CLYBL are routinely utilized for targeted transgene integration in human cells. Recent advances in genome architecture, gene regulation, and genome editing technologies are driving the discovery of novel SHSs for precise and safe genetic modification. This review aims to provide a comprehensive overview of SHSs and their applications that will guide investigators in the choice of SHS, especially when complementary sites are needed for more than one transgene integration. First, it outlines safety and functional criteria that qualify a genomic site as a safe harbor site. It then discusses the two primary strategies for identifying SHSs: i) traditional lentiviral-based random transgenesis, and ii) modern genome-wide in silico screening followed by CRISPR-based validation. This review also provides an updated catalogue of currently known SHSs in the human genome, detailing their characteristics, uses, and limitations. Additionally, it discusses the diverse applications of SHSs in basic research, gene therapy, CAR T cell-based therapy, and biotechnological production systems. Finally, it concludes by highlighting challenges in identifying universally applicable SHSs and outlines future directions for their refinement and validation across biological systems.

1. Introduction

The human genome consists of approximately 3.2 billion base pairs harboring 20,000-25,000 protein-coding genes that make up approximately 1–2% of the entire genome [1,2]. This means that 98–99% of our genome is noncoding and has traditionally been referred to as “junk DNA”, presumed to have only a structural function [3]. However, recent research has revealed that around 80% of the human genome contains functional elements [4]. This includes over 2600 microRNAs (miRNAs), thousands of long non-coding RNAs (lncRNAs), and several classes of transfer RNAs (tRNAs), all playing significant roles in gene regulation [4,5]. Other non-coding functional elements like enhancers, silencers, and promoters orchestrate precise gene expression [6,7]. Repetitive DNA sequences, including short tandem repeats (STRs), contribute to genome variability and can influence gene expression and stability [8]. This uneven distribution of genes and regulatory elements across the genome presents challenges for random transgene insertion, emphasizing the need for precise genome-editing tools to avoid disrupting critical regulatory networks, particularly in genetic engineering and therapeutic contexts [9].
Traditionally, transgene integration has been achieved using retroviral or lentiviral vectors, as well as transposon-based systems such as Sleeping Beauty and PiggyBac, to stably incorporate genetic elements or therapeutic genes into host genomes [10,11]. However, these methods suffer from significant drawbacks. The random integration of transgenes into the host genome poses a significant risk of insertional mutagenesis and disruption of endogenous genes or regulatory elements, potentially leading to oncogenic transformation [12,13,14,15]. Additionally, the integration near regulatory regions can induce transgene inactivation or result in unintended physiological consequences [16]. Furthermore, variations in transgene integration sites among different transgenic cells can also confound phenotypes [17,18]. These drawbacks raise significant concerns about the safety, stability, and reproducibility of random transgene integration methods.
To address these limitations, the concept of Safe Harbor Sites (SHSs), specific loci within the genome that permit stable integration and expression of exogenous genetic material without disrupting native gene function or compromising genomic integrity, has become a cornerstone in the fields of genetic engineering and gene therapy [19,20]. SHSs are typically located in transcriptionally active regions to support the expression of integrated transgenes, but without interfering with the transcription of endogenous genes or increasing the risk of oncogenic transformation [21]. Identification of SHSs has been driven by a combination of computational predictions, empirical studies, and high-throughput screening techniques [22,23]. One of the well-characterized SHSs is the AAVS1 locus on chromosome 19, initially identified as a hotspot for the site-specific integration of adeno-associated virus type 2 and frequently targeted for therapeutic gene integration [24,25,26]. The ROSA26 locus in mice is another well-known and ideal SHS, valued for its permissive and predictable expression profile, together with the absence of a deleterious phenotype when targeted [27,28].
The advent of precise genome-editing technologies, such as CRISPR-Cas9 and TALENs, has greatly enhanced the precise insertion of genetic material into these SHSs, significantly improving the efficiency and reliability of genetic engineering efforts [29,30]. Transgene expression from SHSs ensures controlled and predictable outcomes, mitigating the risks associated with random integration [21,31,32]. Consequently, SHSs provide a robust platform for gene function studies and disease modeling [33], generation of chimeric antigen receptor (CAR) T cells and/or induced pluripotent stem cells (iPSCs) for cell-based therapies [34,35,36], and for the construction of complex genetic circuits with novel functionalities [37]. They also support biopharmaceutical production and the development of genetically modified crops with improved traits while minimizing ecological and biosafety risks [21,35,38].
Despite their advantages, the use of SHSs faces challenges. These include difficulties in identifying universal sites across cell types and tissues due to genomic variability among individuals and species [39]. Risks of off-target effects and unintended alterations during the integration process are possible [40]. Also, the current incomplete annotation, with many genomic regions still poorly characterized and their functional contributions to phenotypic traits still constitute a significant limitation in the use of SHSs [41]. Nonetheless, rapid advances in genome annotation, epigenomics, and single-cell sequencing are enabling the discovery and refinement of SHSs, broadening their biomedical and biotechnological applications [23,42]. Overall, SHSs address limitations of classical transgene integration and open new opportunities in research, therapy, and industry. As our understanding of the human genome deepens and genome editing technologies evolve, SHSs will play a key role in precision medicine and biotechnology. In this review, we provide a comprehensive overview of selection and identification criteria for novel SHSs and the strategies followed to identify them. We discuss in detail the best-characterized SHSs, including those recently discovered, and highlight their applications in fundamental research, gene therapy, and biopharmaceutics production.

2. Criteria of SHS Selection

For a genomic site to qualify as a safe harbor, it must fulfill specific criteria to ensure that transgenic elements are stably expressed in the host genome and do not cause alterations such as transcriptional dysregulation of endogenous genes, oncogenes activation and/or tumor-suppressing genes inactivation [43,44]. These criteria have undergone significant revisions in response to advances in our understanding of genome architecture and function. Based on the genomic data and experimental observations obtained from lentiviral vector integration sites, Sadelain and colleagues (2011) proposed five criteria for a candidate site to qualify as a SHS (Figure 1). First, SHSs must be located at a safe distance, typically at least 50 kilobases (kb), from the 5’ end of coding genes in order to prevent the disruption of essential genes that could lead to cellular dysfunctions [45,46]. Second, SHSs must be at least 300 kb away from cancer-associated genes to minimize the risk of oncogene activation [45,46]. Third, they must be at a distance of at least 300 kb from microRNAs [45,46]. Fourth, they must reside outside of the transcriptional units to avoid insertional mutagenesis or transcriptional dysregulation [45,46]. Fifth, SHSs should be outside of ultra-conserved genomic regions, telomeres, and centromeres to avoid interfering with elements critical for gene regulation [45,46].
While these criteria are based on linear genomic features and provide a foundational framework for SHS identification, it is now clear that the genome is organized into three-dimensional (3D) topologically associated domains (TADs) and gene-gene interactions are often confined within these TADs [47,48]. Therefore, the Sadelain group subsequently suggested that SHSs should not be in TADs enriched in oncogenes or regulatory hotspots to provide additional safety against transcriptional activation of proto-oncogenes [21] (Figure 1). However, long-range interactions between genes that are 500 kb away have been reported, underscoring the need for more cautious site selection [15,49]. Moreover, the ever-increasing knowledge about regulatory non-coding RNAs, including long non-coding and small RNAs, now dictates the exclusion of sites that might disrupt these elements [21,50,51]. Finally, in the selection of SHSs, it must be considered that active chromatin characterized by histone modifications, such as H3K4me1 and H3K27ac, favors robust gene expression [52,53]. Therefore, SHSs should be located in the active chromatin regions to allow sustainable transgene expression, possibly across cell types and conditions [21,23] (Figure 1).
The integration of advanced genomic and chromatin epigenetic landscape data into SHS selection criteria introduces both opportunities and challenges. The fine-tuning of SHS definition for specific therapeutic applications enhances safety, but also reduces its generalizability across diverse contexts [45]. As genome-editing technologies like CRISPR-Cas9 evolve, criteria must also continually adapt to account for integration efficiency, off-target effects, and possible chromatin restructuring [54,55]. The efficiency of genomic integration is crucial for clinical applications, where therapeutic success often depends on the expression levels of the therapeutic gene [56]. Off-target mutations could lead to involuntary consequences, including insertional mutagenesis or interruption of essential genes [57,58]. Similarly, targeting SHSs should not significantly alter cell transcriptomics, proteomics, and metabolomics profiles and should maintain iPSCs’ pluripotency, karyotypes and differentiation capacity [19]. Advances in computational modeling, single-cell transcriptomics, and epigenomics hold promise for refining SHS selection criteria [59]. The ultimate goal is to identify universal SHSs enabling robust transgene expression across diverse tissues, cell types, and physiological conditions (Figure 1, BOX1).
Despite these refinements, the criteria for identifying and validating SHSs remain imperfect for several key reasons (Figure 1, BOX2). First, the annotation of the human genome is still incomplete. Initiatives such as GENCODE have remarkably advanced our understanding of the human transcriptome and non-coding elements, yet many functional genomic regions are still poorly characterized [60,61]. Thus, targeting of SHSs selected based on current annotations could inadvertently disrupt unknown regulatory elements. Second, the dynamic and context-dependent nature of gene regulation complicates SHS selection. In fact, targeting sites deemed safe in one cellular context may cause genetic dysregulation in another due to cell-type-specific enhancers, repressors, or chromatin states. Loci deemed non-essential in certain cell types could still have significant biological roles at the systemic level [62,63]. Epigenetic modifications are highly variable and may change over time or in response to environmental cues, leading to alterations in gene expression. This variability may affect the stability and efficacy of transgene expression, posing challenges for long-term predictability [64,65]. Finally, the role of chromatin topology in defining SHSs is not fully understood. In fact, while TAD boundaries and insulator elements are increasingly recognized as critical regulatory structures, the precise mechanism and extent of their influence on gene expression across various cell types are still under investigation [66,67].

3. Identification and Validation of SHSs

Identification and characterization of SHSs are fundamental for advancement of genetic engineering, genome editing and gene therapy [68]. Two different but complementary strategies have been developed for novel SHS identification: a classical experimental approach and a computational (i.e., in silico) method [45,51,68,69]. The classical approach primarily relies on the use of retroviral/lentiviral vectors, which integrate semi-randomly into the host genome, preferentially targeting transcriptionally active regions [70]. This inherent bias can be leveraged to identify genomic loci that support stable transgene expression [71,72]. The classical approach involves the optimization of transduction conditions to favor single-copy integrations per cell/genome. The validation of reliable transgene expression includes the use of RT-PCR, flow cytometry, immunofluorescence, and Western blots [45]. This is followed by mapping of integration sites in clones with stable expression, performing inverse and/or ligation-mediated PCR and DNA sequencing [73,74,75]. Finally, candidate loci are evaluated against the SHS safety criteria discussed above to exclude sites occurring in proximity of cancer-related genes, and/or those inside a transcriptional unit that can cause critical alterations in global gene expression as assessed by transcriptomics and proteomics profiling, or in cell karyotype, pluripotency, and differentiation capacity [45,75] (Figure 2). Following this approach, Papapetrou et al. (2011) identified a SHS located in chromosome 1 that met all five foundational criteria and achieved stable β-globin expression in iPSCs derived from thalassemia patients without perturbing neighboring gene expression [45].
The classical approach can also make use of site-specific integrases or homing endonucleases that have numerous pseudo-sites in the human genome. Liu et al. (2009) employed phiC31 integrase to target a GFP reporter cassette into pseudo-attP sites within the human genome, identifying a locus on chromosome 13q32.3 that supported long-term expression in human embryonic stem cells (hESCs) and during neuronal differentiation [76]. Overall, the classical approach enables direct identification and functional validation of candidate SHSs within specific biological contexts and can uncover previously unrecognized loci suitable for safe transgene integration [45,77]. However, it is resource-intensive, requiring careful optimization of transduction conditions and extensive screening to isolate suitable clones that meet the SHS safety criteria and support sustainable gene expression. Additionally, the classical approach is limited by its dependence on the chromatin state of the screened cell type, which may lead to the identification of SHSs that are not active across different cellular contexts [45].
The computational in silico approach employs genome-wide bioinformatics-guided strategies to identify candidate SHSs using genomic and epigenomic datasets [23,51,69]. This strategy typically begins with systematic screening of the genome to locate regions devoid of essential genes, cancer-associated genes, and regulatory elements while favoring integration sites within transcriptionally active chromatin to ensure sustainable transgene expression [23,44,51]. Once candidate loci are identified, the experimental validation starts. Sequence-specific nucleases such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), or CRISPR/Cas9 are employed to introduce double-strand breaks (DSBs) at the target site. A transgene or reporter gene cassette is then integrated at the break site [51,69,78,79] (Figure 2). The correct site-specific integration is confirmed by PCR using primers flanking the genomic region and the transgene cassette [80,81]. This is followed by functional validation of the integrated genes, e.g., reporter genes, using methods such as RT-PCR, flow cytometry, and Western blot, monitored over an extended period of time to evaluate expression stability and potential silencing [51,69] (Figure 2).
In addition to expression analysis, the safety and biological neutrality of the candidate SHSs are assessed through global transcriptomic profiling. Importantly, the impact on iPSC pluripotency status, karyotypes, differentiation potential, and possible silencing upon differentiation are also considered and assessed [43,51,69]. This comprehensive approach was recently used to identify several new SHSs, such as Rogi1 and Rogi2 [51], Pansio-1, Olonne-18, and Keppel-19 [69], and specific SHSs in blood cell lineages [44]. Compared to the classical experimental approach, the in silico method is efficient, cost-effective, and reduces the number of required experimental iterations by pre-screening genomic regions using computational tools. It can also be applied across various organisms and cell types, provided that high-quality genomic and epigenomic datasets are available. However, computational predictions must be experimentally validated, as they may not fully account for complex biological systems and are limited by the accuracy and resolution of current genome annotations and epigenomic maps.

4. Known Genomic Safe Harbors: Features and Limitations

4.1. AAVS1

The Adeno-Associated Virus Site 1 (AAVS1), located on chromosome 19q13.4, was originally identified as the preferential genomic integration site for adeno-associated virus type 2 (AAV2) [24,82,83,84]. Integration at AAVS1 is mediated by the AAV Rep protein, which recognizes specific sequence motifs within the site that resemble the viral origin of replication [82,85]. In the absence of Rep, AAV vectors tend to integrate randomly or persist predominantly as episomal DNA [46,86]. AAVS1 resides within intron 1 of the PPP1R12C gene, which encodes a regulatory subunit of myosin protein phosphatase [87]. While PPP1R12C (Protein Phosphatase 1 Regulatory Subunit 12C) is ubiquitously expressed across various human tissues and primary cells, its specific biological function remains incompletely characterized [87]. As SHS, earlier investigations showed that AAVS1 allowed efficient transgene expression in hESCs and iPSCs, and their differentiated progeny without impairing pluripotency and differentiation capacity [88,89]. Subsequent work extended its use to various cell lines, including K562, HeLa, A549, HEK293, U2OS, and mesenchymal stem cells, for applications such as transgene expression and shRNA-mediated gene silencing [90,91,92]. With the advent of CRISPR-Cas9 technology, targeting AAVS1 for transgene expression has become a routine laboratory tool for gene function studies, disease modeling, and therapeutic gene delivery [93,94,95,96,97]. However, emerging evidence indicates that AAVS1 targeting is not without limitations. Silencing and/or expression variability of genes integrated at this locus has been observed in iPSC-derived cardiomyocytes [98,99], endothelial cells [98], myeloid cells [100], hepatocytes [101] and Jurkat T cells [102]. Additionally, transgene integration at AAVS1 perturbed endogenous gene expression, including PPP1R12C [103,104], and neighboring genes TNNI3 (Troponin I3) and PPP6R1 (Protein Phosphatase 6 Regulatory Subunit 1) [23]. Furthermore, given that AAVS1 lies in a gene-dense genomic region, the potential for unintended dysregulation of additional neighboring genes remains underexplored and requires further investigation (Table 1).

4.2. hROSA26

The human homolog of murine Rosa26 locus (hROSA26) was identified by searching the human genome using the mouse ROSA26 transcript as a template [27,105]. The search resulted in a region with high sequence similarity (85%) on human chromosome 3 (3p25.3) in the intron 2 of THUMPD3 (THUMP domain 3 tRNA Guanosine Methyltransferase) gene, and hROSA26 mRNA was detected in various hESCs and tissues [27,106]. THUMPD3, in complex with TRMT112 (tRNA methyltransferase activator subunit 11-2), functions as an N2-methylguanosine (m2G) methyltransferase that modifies specific residues in a broad range of cytoplasmic tRNAs, thereby playing a significant role in translation regulation and cellular growth [107]. Targeting the hROSA26 locus with red fluorescent protein (RFP) reporter cassette permitted stable expression in hESCs [27]. Subsequent recombinase-mediated cassette exchange (RMCE) successfully replaced RFP with a puromycin resistance gene, demonstrating the locus’s amenability to efficient and versatile genome engineering [27]. Importantly, hESCs targeted at the hROSA26 locus retained typical stem cell morphology, alkaline phosphatase activity, normal karyotype, and the capacity to differentiate into neurons, chondrocytes, smooth muscle cells, and hepatocytes [27,108]. They also preserved the capacity to differentiate into the three germ layers, ectoderm, endoderm and mesoderm, and trophoblast lineage [27]. Similarly, hiPSCs targeted at the hROSA26 locus were differentiated into microglia-like cells (MGLs), which exhibited canonical morphological, transcriptional, and functional properties of native microglia in monoculture, co-culture, and 3D organoid models [109]. The locus has also been utilized for stable transgene expression in various human cell lines, including hTERT-RPE1 (hTERT-immortalized Retinal Pigment Epithelial 1) [110], and HEK293 (Human Embryonic Kidney 293) cells [111,112]. Collectively, these findings establish hROSA26 as a locus suitable for stable and consistent transgene expression across multiple human cell types, making it a valuable platform for genetic engineering and biomedical research. Nevertheless, this locus, similar to AAVS1, is situated in a gene-rich region, raising concerns about potential disruption of nearby genes upon transgene integration [45]. Furthermore, since the locus lies within intron 2 of THUMPD3, a gene whose function is not yet fully understood, the long-term consequences of its perturbation remain unclear (Table 1). A comprehensive transcriptomic analysis following hROSA26 targeting is currently lacking, and further investigation to assess the broader impact of genetic manipulation at this locus is needed.

4.3. H11 Locus

The human Hippo 11 (H11) locus is located on chromosome 22q12.2 within a transcriptionally active intergenic region flanked by the DRG1 (Developmentally Regulated GTP Binding Protein 1) and EIF4ENIF1 (Eukaryotic translation Initiation Factor 4E Nuclear Import Factor 1) genes. It was identified through comparative genomic analysis using the murine orthologs of DRG1 and EIF4ENIF1, which revealed a conserved gene arrangement and intergenic distance between the two genes in both species, despite only ~45% sequence identity and absence of highly conserved elements [113]. This locus is situated approximately 4500 and 683 bp downstream of DRG1 and EIF4ENIF1, respectively, as both genes are transcribed from the opposite strands [113]. The H11 locus fulfills certain safety criteria, including absence of known oncogenes and microRNAs within a 300 kb window while residing in a region with high transcriptional activity [113]. Targeting this locus supported stable expression of various transgenes in hiPSCs, including transcription factors (LMX1A, FOXA2, and OTX2), the auxin-inducible degron component TIR1, and coagulation factor VIII, even after extensive passaging and differentiation, with no evidence of transgene silencing [113,114,115] (Table 1). Additionally, the H11 locus allowed recombinase-mediated cassette exchange (RMCE) using phiC31 and Bxb1 integrases in both healthy and patient-derived hiPSCs [113,115]. Importantly, targeted hiPSC cells maintained typical pluripotent morphology, expressed canonical pluripotency markers, preserved normal karyotypes, and readily differentiated into derivatives of all three germ layers (endoderm, mesoderm, and ectoderm), confirming their intact developmental potential [113,114]. In summary, the H11 locus represents a promising intergenic SHS for stable transgene integration, supporting high and consistent expression without impairing stem cell pluripotency or differentiation capacity. However, the effect of targeting this locus on global transcriptomics profiling and the expression of neighboring genes is not known and requires further investigation.

4.4. CCR5 Locus

CCR5 (C-C motif chemokine receptor 5) gene, located on chromosome 3p21.31, encodes for a receptor involved in the immune response [116]. CCR5 is best known for its role as a co-receptor facilitating Human Immunodeficiency Virus (HIV) entry into host cells [116]. The interest in CCR5 as a potential SHS site for transgene integration stemmed from studies of individuals carrying a 32-base pair deletion (CCR5-∆32). This naturally occurring mutation renders CCR5 non-functional and confers resistance to HIV infection without causing significant adverse effects [117,118,119]. These findings suggest that CCR5 can be disrupted without detrimental consequences, making it an attractive candidate for targeted transgene integration. Experimental disruption of the CCR5 gene in hiPSCs did not alter the expression of pluripotency markers or impair differentiation capacity into the three germ layers and CD34+CD43+ hematopoietic progenitors, and maintained a normal karyotype [120,121]. The ZFN-mediated GFP gene integration into the CCR5 locus in hESCs resulted in stable expression without affecting their pluripotency or differentiation capacity into neural progenitors [122]. Furthermore, chimeric antigen receptor (CAR) genes integrated into the CCR5 locus in T cells enabled stable transgene expression, conferred them with resistance to HIV infection, while allowing these modified T cells to target HIV-infected cells [123]. Despite its potential as a SHS, certain challenges have been encountered when using the CCR5 locus for transgene expression, including inherently low expression likely due to heterochromatin markers and/or gradual decline in transgene expression from CCR5 [35,124,125]. Targeting the CCR5 locus has also been associated with upregulation of neighboring genes, including CCR1 and CCR3 [125]. Importantly, disruption of CCR5 increases the susceptibility to West Nile virus infection that can cause fatal encephalitis [126,127] (Table 1).

4.5. Citrate Lyase Beta-Like (CLYBL)

CLYBL locus, mapped to intron 2 of the Citrate Lyase Beta-Like (CLYBL) gene on the long arm of chromosome 13 (13q32.1), was initially identified as a potential SHS site through phiC31 integrase-mediated targeting of a GFP reporter into a pseudo-attP site in this region [128]. The CLYBL gene encodes citramalyl-CoA lyase, a ubiquitously expressed mitochondrial enzyme with malate/β-methylmalate synthase activity, involved in the C5-dicarboxylate metabolic pathway, including itaconate metabolism. Loss of CLYBL gene function has been associated with vitamin B12 deficiency [129,130]. Transgenes integrated into the CLYBL locus in hiPSCs and neuronal stem cells (NSCs) yielded stable and superior expression to that achieved from the AAVS1 site [104] (Table 1). Importantly, the transgene integration at the CLYBL locus caused less disruption to local gene expression compared to other loci. For instance, while targeted transgene integration into the AAVS1 locus showed significant downregulation of the PPP1R12C gene (>1000-fold) and upregulation of other neighboring genes, targeting the CLYBL locus resulted in only a ~50-fold decrease in CLYBL expression with minimal impact on neighboring gene activity [104]. Numerous studies have shown that targeted gene integration into CLYBL site of hESCs and iPSCs not only supports a strong transgene expression but also preserves the parental karyotype, typical stem cell morphology, expression of pluripotency markers (Oct4, SSEA4, Tra1-60, and Tra1-8), while maintaining the capability to differentiate into various lineages (e.g., neurons, microglia-like cells, astrocytes) and to form all three germ layers [104,128,131,132,133,134,135]. The functional utility of this locus has also been demonstrated in HeLa and HEK293 cells [135]. This locus has also successfully been targeted using exchangeable DNA payloads, enabling insertion of various transgenes for diverse research applications [136]. These findings underscore the CLYBL locus’s potential as a valuable site for safe and efficient transgene integration, particularly in the context of stem cell research and therapeutic applications. Its favorable genomic context and minimal impact on surrounding gene expression enhance its appeal as a safe locus. However, the long-term stability of transgene expression and the impact on the global transcriptomics profile upon CLYBL targeting remain to be assessed.

4.6. SHS231

The homing endonuclease mCre1 is a class of enzymes that recognize and cleave a specific 20 bp DNA sequence to catalyze the lateral transfer of parasitic DNA elements [137]. Comprehensive studies have demonstrated that mCreI can still cleave DNA sites with a single base substitution (19/20 bp identity, or 95%) at approximately 90% efficiency [137,138,139]. Pellenz and colleagues searched the human genome for potential mCreI pseudo-sites, including those with one base-pair substitution (19/20 bp) [80,140]. This analysis identified 35 sites across 16 chromosomes, including both arms of the X chromosome, and in vitro studies confirmed that mCreI could cleave these sequences [80]. These candidate sites were evaluated using eight SHS criteria to assess safety, functionality, and structural accessibility and were compared to the established SHSs, i.e., AAVS1, CCR5 and hROSA26. Notably, ten of the mCreI target sites met 6–7 of the eight core SHS criteria, whereas AAVS1, CCR5, and hROSA26 met only 5, 5, and 3 criteria, respectively [80]. One promising site, designated as SHS231, located on the long arm of chromosome 4, met 7 out of the 8 SHS criteria. This site was targeted using CRISPR-Cas9 for both homology-dependent and non-homology-dependent transgene integration in various rhabdomyosarcoma (RMS) cell lines (Rh5, Rh30, and SMS-CTR) as well as in HEK293T cells [80]. Targeting the SHS231 site enabled GFP expression for more than 45 days in Rh5 and SMS-CTR cells, indicating robust integration and stable transgene expression [80]. Additionally, when targeted with a Cas9 expression cassette, the SHS231 site supported stable Cas9 expression, enabling functional genome editing, for instance, (i) Cas9 mediated a 17 kb deletion in the PAX3/FOXO1 fusion oncogene in Rh30 cells, and (ii) Cas9-VPR integrated at SHS231 activated the MYF5 promoter in RMS cells, inducing robust expression [80]. These findings suggest that SHS231 is a novel and promising SHS that can be targeted in various human cell lines for application in gene therapy, functional genomics, and disease modeling [80] (Table 1). More recently, Vlassis et al. demonstrated that four additional sites identified in the Pellenz study, namely SHS257 (chr7), SHS325 (chr8), SHS313 (chrX), and SHS301 (chr7), also enabled efficient and stable expression in hiPSCs, primary T and natural killer (NK) cells and in Jurkat cells [141]. These sites supported sustained transgene expression throughout the differentiation of iPSC toward CD34+ hematopoietic stem and progenitor cells, lymphoid progenitor cells, and NK cells [141]. However, additional studies are required to assess global transcriptomic alterations and the long-term stability of transgene expression upon targeting these novel SHSs.
Table 1. A summary of SHSs in human genome and their features.
Table 1. A summary of SHSs in human genome and their features.
SHS LocusChrom-
osome
Intergenic/
Intragenic
Cell ModelsTransgene ExpressionEffect on Stem Cells CharacteristicsEffect on TranscriptomicsReported
Silencing
Gaps and LimitationsRefs
AAVS119q13.42Intragenic
PPP1R12C
Many cellular models, including hESCsRobust expressionNo effect on hESCs’ pluripotency and differentiation capacityUnknownYesLocated within PPP1R12C
transcriptional unit and in a gene-dense region
[87]
hROSA263p25.3Intragenic
THUMPD3
Many cell models, including hESCsRobust expression No effect on hESCs’ pluripotency and differentiation capacityUnknownNo Located within THUMPD3 gene transcriptional unit and in a gene-dense region[27]
H1122q12.2Intergenic
DRG1-H11-EIF4ENIF1
Mainly in hiPSCsRobust
expression
No effect on hESCs’ pluripotency and differentiation capacityUnknownNoLocated between two genes whose dysregulation upon targeting is not known[113,114,115]
CCR53p21.31Intragenic CCR5 genehESCs, T cells and HEK293T cellsLow
expression
No effect on hESCs’ pluripotency and differentiation capacityUnknownYesUpregulated the flanking genes CCR1 and CCR3, and increased the susceptibility to West Nile virus infection[122]
CLYBL13q32.1Intragenic
CLYBL gene
iPSCs, hESCs, NSC and in Hela and HEK293 cellsRobust
expression
No effect on hESCs’ pluripotency and differentiation capacityUnknownNoLocated within coding gene, and the long-term transgene expression is not known[104,132]
SHS2314q13.1IntergenicRh5, Rh30, SMS-CTR, and HEK293TRobust expressionUnknownUnknownNoTransgene expression in other cell models, like iPSCs, is not assessed[139]
Rogi11q31.3IntergenicHEK293T, Jurket, and primary T cells and iPSCsRobust expressionUnknownMinimal effectYesThe effect on stem cells’ pluripotency, karyotype, and differentiation capacity is not assessed[51]
Rogi23p24.3
Pansio-11p13.2IntergenichESCs and iPSCsStable expression No effect on hESCs’ pluripotency and differentiation capacityMinimal effectNoThe long-term stability and transgene expression in other cell models are not assessed [69]
Olonne-18 18q21.31Intergenic
Keppel-1919p13.3Intergenic
BLD_SHS103p22.2Intronic
GOLGA4
HUDEP2 erythroid progenitorsStable expressionUnknownUnknownNoThese sites may be targeted in HSCs to assess the impact on HSC physiology and differentiation capacity into diverse blood cell lineages.[23]
BLD_SHS146q25.3Intronic
ARID1B
BLD_SHS158q24.12Intronic/
TAF2
eSHS67q36.1Pseudogene ZNF767PT cellsStable expression UnknownUnknownNoFunctionality of this locus in other blood cells and iPSCs warrants further assessment.[44]

4.7. Rogi1 and Rogi2

Aznauryan and colleagues employed a computational approach to identify novel SHSs suitable for safe transgene integration and stable expression [51]. Their strategy excluded sites located in gene-dense regions, or within transcriptional units, and those near oncogenes, microRNAs, long non-coding RNAs, tRNAs, or transcriptional enhancers. Additionally, candidate sites were also required to be at least 300 kb away from telomeres and centromeres [51]. This stringent filtering process yielded over 2000 potential SHSs distributed across the human genome that satisfied the defined safety criteria. Among these, two sites named as Rogi1 and Rogi2 (Region for optimal gene insertion 1 and 2) localized on chromosomes 1 and 3, respectively, were experimentally validated as SHSs. Both sites enabled robust and sustained expression of the reporter gene mRuby for over 90 days in HEK293T and Jurkat cell lines, outperforming traditional SHSs such as AAVS1 and CCR5 [51] (Table 1). Similarly, in human primary T cells, integration into Rogi1 and Rogi2 supported stable mRuby expression for over 20 days. In human dermal fibroblasts, expression of the therapeutic gene LAMB3 from both sites remained sustained for at least 25 days [51]. Transcriptomic analysis of HEK293T and Jurkat cells harboring mRuby at the Rogi2 locus revealed a preserved global transcriptional profile [51]. Similarly, integration of mRuby into Rogi1 in primary human T cells resulted in minimal transcriptional alteration, as evidenced by single-cell RNA-seq analysis. Importantly, no cancer-related pathways were upregulated; in fact, the JUN oncogene, which encodes the c-Jun transcription factor, was downregulated [51]. Of note, reporter gene expression displayed clonal heterogeneity in HEK293T and Jurkat cell lines, which is likely attributable to the inherent genomic instability of immortalized cancer cells [51]. A recent study targeted the transcription factor FOXN1 gene into the Rogi1 site in iPSCs and subsequently differentiated them into thymic epithelial cells upon FOXN1 induction [142]. However, this work reported the silencing of the tetracycline repressor and leakage of FOXN1 expression, which were mitigated by introducing an A2-ubiquitous chromatin opening element (A2UCOE) and an SV40 poly(A) termination sequence upstream of the tetracycline-inducible promoter [142]. It remains unclear whether this silencing and leakage are attributable to the genomic locus used or are intrinsic properties of the cassette. Furthermore, the effects on pluripotency, karyotype, and differentiation potential are not widely assessed yet and warrant further investigation.

4.8. Pansio-1, Olonne-18, and Keppel-19

Autio and colleagues employed a computational strategy to identify new SHSs in the human genome that fulfill the established SHS selection criteria [69]. The analysis resulted in the identification of 25 putative SHS loci. Three of them-Pansio-1, Olônne-18, and Keppel-19-were experimentally validated in hESCs (Table 1) [69]. Targeted transgene integration at these loci had a minimal effect on the native transcriptomic profiling and caused no karyotype or pluripotency abnormalities [69]. Furthermore, targeting these sites did not impair the iPSCs’ capacity to differentiate into neuronal, liver, cardiac, and pancreatic β-cells or to give rise to endoderm, mesoderm, and ectoderm tissues [69]. Importantly, the expression of MAGI3, TXNL1, and ZNRF4 genes closest to Pansio-1, Olônne-18 and Keppel-19, respectively, was not significantly altered following transgene integration. Additionally, the transgene expression from these loci remained stable for at least 15 passages and was maintained following differentiation into neuronal, hepatic, and cardiac lineages [69]. Notably, these loci enabled sustainable transgene expression both from constitutive and inducible (Tet-On) promoters integrated into these sites and supported Bxb1 integrase-mediated cassette exchange [69,143]. While these findings highlight the potential of Pansio-1, Olônne-18, and Keppel-19 as valuable SHS sites, further research is needed to assess long-term transgene stability, validate these loci across additional cell types, and confirm their performance in in vivo models.

4.9. Cell/Tissue Specific SHSs

Traditional approaches for identifying SHSs have primarily focused on static genomic features, often overlooking tissue-specific genome architecture, transcriptional activity, and regulatory elements [51,69,144]. Incorporating 3D chromatin organization in a tissue-specific manner into SHS identification methods can help select a more refined, cell-type-specific SHS that supports stable transgene expression without disrupting essential genes or interfering with native transcriptional programs [23]. Shrestha et al. analyzed polymorphic mobile element insertion sites, epigenomic signatures, and 3D chromatin structure using data from the 1000 Genomes Project and the Genotype-Tissue Expression project. Their aim was to identify loci where mobile elements had inserted without detrimental effects, suggesting potential tolerance for additional genetic material [23]. This integrative approach led to the identification of 19 unique SHSs in blood cells (designated as BLD_SHS1-BLD_SHS19) and 5 in brain cells (designated as BRN_SHS1-BRN_SHS5). Most of these sites were located in the intronic regions, and only one (BLD_SHS_7) was found in an intergenic region. All SHSs were mapped into active chromatin regions, and 13 were located outside of TADs with high gene-density or in TADs harboring cancer-related genes, particularly in the lymphoblastoid GM12878 cell line. Among these, BLD_SHS10 (chr3:37361602–37361603) resides in the intron of the GOLGA4 gene, which is the sole gene within its relative TAD [23]. Three candidate sites, BLD_SHS10, BLD_SHS14, and BLD_SHS15, were experimentally validated in HUDEP2 erythroid progenitors, demonstrating that transgenes integrated at these sites exhibited stable expression over several weeks and after differentiation into mature erythrocytes, without significantly disrupting the expression of nearby genes [23] (Table 1). These findings indicate that the newly identified loci may serve as tissue-specific SHSs for therapeutic gene integration, potentially enhancing the safety and precision of gene and cell-based therapies [23].
Odak et al. identified a novel extragenic SHS site that can be used for precise therapeutic T-cell engineering. They employed a computational and experimental framework to identify and validate SHSs in human peripheral T cells that satisfy key safety and efficiency criteria [44]. Specifically, the assay for transposase-accessible chromatin coupled to high-throughput sequencing (ATAC-seq) was used to identify regions of open chromatin in T cells, revealing 379 candidate sites characterized by strong ATAC-seq peaks and that fulfilled SHSs safety criteria [44]. Among them, an extragenic site designated as eSHS6 localized to chromosome 7, within the pseudogene ZNF767P, showed high cleavage efficiency (>80% when targeted with CRISPR/Cas9) and chromatin accessibility [44] (Table 1). Human peripheral blood T cells were targeted with CRISPR/Cas9 and a donor vector to integrate a CD19-CAR construct into the eSHS6 site and then evaluated in vitro and in vivo for CAR expression and T-cell function [44]. The resulting eSHS6-CAR T cells maintained durable CAR expression over multiple weeks of antigen stimulation and provided long-term leukemia control in vivo [44]. eSHS6-CAR T cells maintained robust anti-tumor activity even after multiple leukemia rechallenges and demonstrated proliferation, persistence, and tumor clearance similar to T-Cell Receptor Alpha Constant (TRAC)-CAR T cells [44]. Noticeably, eSHS6-CAR T cells, unlike TRAC-CAR T cells, retained their native T-cell receptor, which could be beneficial for additional antigen recognition and expansion [44]. Thus, this study represents the first systematic identification and functional validation of extragenic SHSs in human T cells, highlighting eSHS6 as a promising alternative to TRAC for CAR integration for safer and more versatile T-cell-based immunotherapies [44].

5. Safe Harbor Sites Applications

5.1. SHSs’ Uses in Basic Research

Targeted integration of genetic material into well-characterized SHSs offers a powerful strategy for achieving controlled and predictable gene expression for functional studies, without disrupting endogenous gene function [21,145]. Unlike traditional lentiviral methods, which are prone to random insertion and position-dependent variation, SHS-targeted integration provides consistent transgene expression, thereby enhancing the reliability and reproducibility of experimental outcomes [20]. This approach has enabled a wide range of innovative applications. For instance, the SHS for transgene expression in iPSCs is used for the generation of 3D organoid models to accurately study complex biological processes [145]. Indeed, human iPSCs engineered to express channelrhodopsin-2 (ChR2) from the AAVS1 locus and differentiated into forebrain organoids exhibited a robust neural activation upon light stimulation [87]. When these organoids were connected with spinal cord and skeletal muscle organoids, they triggered consistent muscle contractions, offering a robust model to study brain function and brain-periphery neural circuits [87]. Similarly, Zhang et al. targeted hiPSCs at the AAVS1 locus with a voltage-sensitive fluorescent protein, enabling real-time action potential recordings in differentiated ventricular, atrial, and nodal cardiomyocytes in both 2D and 3D cultures. This non-invasive approach provides a scalable alternative to traditional patch-clamp techniques for electrophysiological studies [146]. Integration into SHSs can also be used for the creation of reliable disease models. Dost et al. engineered hiPSCs to express the oncogenic KRASG12D from the AAVS1 locus, generating an early-stage lung adenocarcinoma organoid. These organoids showed a loss of differentiation markers in alveolar type 2 cells upon KRAS activation, mimicking both mouse models and human tumor samples [147]. SHSs can also be used to integrate lineage-specific transcription factors in hiPSCs, which can be induced to facilitate direct differentiation into various cellular lineages such as neurons and myoblasts, avoiding the use of cost- and time-consuming protocols. In this regard, human iPSCs targeted at the AAVS1 and CLYBL loci with an open reading frame encoding the transcription factor PAX7 were readily differentiated into expandable myogenic progenitors capable of robust engraftment upon induction of PAX7 expression [148,149]. Also, hiPSCs targeted with the neuronal transcription factor neurogenin 2 at the AAVS1 locus differentiated into mature neurons upon the induction of neurogenin 2 expression [150,151,152,153]. SHS transgene integration also enables the generation of cellular models that can be used for functional genomics, gene-gene interaction, gene dependency mapping, and high-throughput drug screening studies [108,154]. Landin et al. developed hiPSCs and mouse models expressing CRISPR/Cas9 from the AAVS1 and murine ROSA26 loci, respectively [155]. When these mice were transduced with AAV vectors carrying guide RNAs and repair templates specific to KRAS, TP53, or STK11, they developed lung adenocarcinomas that faithfully recapitulated human tumor pathology and drug response, highlighting the model’s translational relevance [155]. In summary, those pioneering studies point to SHSs as invaluable tools for basic research.

5.2. SHSs for CAR T Cell-Based Therapy

CAR T-cell therapy is an immunotherapeutic approach in which a patient’s T cells are genetically engineered to express CARs that selectively recognize and eliminate cancer cells [156]. This personalized treatment harnesses the body’s own immune system to target especially hematologic malignancies such as acute lymphoblastic leukemia (ALL) and multiple myeloma [157]. Ongoing efforts are also exploring its potential in treating solid tumors and infectious diseases [158]. Conventional CAR T-cell production relies on random transgene integration, often resulting in variable and suboptimal expression [159,160]. Targeted insertion of CAR cassettes into SHSs offers a powerful alternative, enabling consistent expression and improved safety profiles [161,162,163,164,165]. Indeed, CAR T-cells engineered to express an anti-CD105 nanobody from the AAVS1 site displayed hallmark features of activated cytotoxic T-cells when co-cultured in vitro with CD105-positive cancer cells: they proliferated, secreted pro-inflammatory cytokines, and efficiently eliminated target cells in vitro. In vivo, these cells markedly inhibited the growth of CD105-positive tumors and prolonged survival in tumor-bearing NOD/SCID mice [166]. Similarly, the CD19-CAR construct targeted to the eSHS6 site mentioned above showed sustainable CAR expression with or without antigen stimulation, effectively lysed CD19+ NALM6 cells, and provided long-term tumor control in an NSG (NOD/SCID/IL-2Rγ null) model of B-cell ALL [44]. In a different study, anti-CD19 CAR cassettes containing 4-1BB and CD3ζ signaling domains were integrated into the AAVS1 site or PD-1 gene locus of T cells. These engineered T cells eradicated tumor cells in xenograft mouse models [162]. Interestingly, in a clinical trial of eight patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma, engineered cells achieved a strong (87.5%) remission rate without severe adverse events even at a low infusion dose [162].
SHSs can also be exploited to engineer immune cells other than T-cells to exert a specific function. For instance, hiPSC-derived neutrophils engineered to express a CAR targeting the prostate-specific membrane antigen from the AAVS1 locus showed a potent cytotoxic effect against the prostate LNCaP cell line in vitro [161]. Similarly, hiPSCs modified to express CARs from the AAVS1 or H11 site were differentiated into CAR macrophages that efficiently phagocytosed tumor cells in vitro and exhibited potent antitumor effects in mouse models of ovarian cancer and neuroblastoma [167,168]. SHS targeting can also be explored for engineering CAR cells to combat infectious diseases. Single-chain variable fragments (scFvs) derived from broadly neutralizing anti-HIV antibodies, integrated into the T cell CCR5 locus, conferred potent lytic activity against HIV-infected cells both in vitro and in a PBMC-humanized mouse model of HIV infection [123]. Similarly, integration of an anti-CD19 CAR into the CCR5 site generated T cells that are resistant to HIV infection while maintaining robust antitumor activity against HIV-associated B-cell malignancies in vitro and in vivo [169]. Altogether, these studies suggest that precise CAR integration into SHSs enhances expression stability, functional potency, and biosafety across diverse immune cell types. This strategy represents substantial advances over random integration, offering greater predictability and durability for CAR-based therapies against both cancer and infectious diseases. However, significant challenges remain in the efficient isolation of primary blood cells, particularly T cells, as well as in their ex vivo expansion and genetic modification, which can affect cell viability, editing efficiency, and long-term therapeutic function.

5.3. Use for Targeted Gene Therapy

Gene therapy using SHSs has emerged as a transformative strategy for treating genetic disorders, cancers, and chronic diseases with significant advantages over traditional approaches using lentiviral vectors [170,171,172,173,174,175,176]. Several proof-of-concept studies highlight the therapeutic potential of SHS targeting. In primary hyperoxaluria type 1 (PH1), insertion of the alanine–glyoxylate aminotransferase (AGXT) gene into the AAVS1 site of PH1 patient-derived iPSCs enabled stable AGXT expression both in iPSCs and differentiated hepatocyte-like cells, suggesting their potential use in autologous cell-based gene therapy for PH1 treatment [174]. In mucopolysaccharidosis type I (MPSI), integration of the iduronidase (IDUA) gene into the CCR5 locus of human CD34+ hematopoietic stem cells (HSCs) led to high levels of the IDUA enzyme secretion, maintained capacity to differentiate into multiple blood cell lineages, and improved both biochemical and phenotypic abnormalities in an immunocompromised MPSI mouse model [177]. In X-linked chronic granulomatous disease, the insertion of the CYBB gene (encoding the gp91phox subunit of NADPH oxidase) into the AAVS1 site of patient-derived CD34+ HSCs restored gp91phox expression and NADPH oxidase activity in ex vivo–derived neutrophils [178]. When these gene-corrected HSCs were transplanted into NSG mice, 4–11% of bone marrow-derived human cells maintained gp91phox expression [178]. Similarly, the integration of coagulation factor VIII (FVIII) into the AAVS1 locus of hemophilia A (HA) patient-derived iPSCs supported stable FVIII expression in their endothelial derivatives. Transplantation with these engineered endothelial cells into an immunocompetent HA mouse model resulted in a 2.12-fold increase in plasma FVIII activity and improved survival following haemorrhagic injury [179]. Similarly, hiPSCs derived from a hemophilia B patient with the targeted insertion of the coagulation factor IX (F-IX) into the AAVS1 locus readily differentiated into hepatocytes capable of stable F-IX secretion and short-term functional activity after engraftment into NOD/SCID mice [180]. Collectively, these studies underscore the potential of SHSs as valuable tools for targeted gene therapy. Nonetheless, the emergence of more precise gene editing technologies like base and prime editors capable of precisely correcting pathogenic mutations will likely limit the use of SHSs for certain gene therapy applications.

5.4. Biotechnology Applications

Pharmaceutical proteins, including hormones, vaccines, enzymes and antibodies, are typically produced using genetically engineered microbial cells or mammalian cell lines like Chinese hamster ovary (CHO) cells and HEK293 cells targeted with transgene cassettes using lentiviral and retroviral vectors [181,182,183,184,185]. SHSs represent now the tool of choice for therapeutic protein expression as they allow robust and stable expression over extended culture periods [124]. In this regard, Deng et al. utilized CRISPR/Cas9 to integrate human serum albumin (HSA) and interferon-β-HSA fusion (IFNβ-HSA) into hROSA26, AAVS1, and CCR5 loci of HEK293T and assessed their stability in high-density suspension culture [35]. The expression from the hROSA26 locus proved the most stable, maintaining > 98% of transgene expression after 60 passages, while that from the CCR5 site showed reduced expression, likely due to transcriptional silencing [35]. The study also demonstrated that the highest yield of HSA and IFNβ-HSA was obtained from hROSA26-engineered cells [35]. Comparable yield was also achieved targeting the AAVS1 locus, whereas insertion at the CCR5 yielded a markedly low amount. Although the obtained yields remain insufficient for industrial use, targeting multiple loci within the same engineered cell line combined with optimized culture conditions may enhance productivity [35]. In this regard, Yang et al. integrated the CTLA4-Ig fusion, made of the cytotoxic T-lymphocyte-associated protein 4 and the IgG1 Fc fragment used for immunosuppression, into the AAVS1 locus of HEK293T cells, achieving stable protein production [186]. Notably, HEK293 cells are advantageous for biopharmaceutical protein manufacturing because they are of human origin: they are able to produce proteins with proper folding and post-translational modifications [187,188]. Also, HEK293 cells are easy to manipulate, they readily domesticate in suspension, and, unlike CHO cells, do not usually produce clipped protein products [187,188]. In summary, SHSs are the tool of choice for biopharmaceutical protein production due to their transgene expression homogeneity and stability. However, both identifying the optimal loci and refining culture conditions remain a critical challenge for achieving industrial-scale protein synthesis.

5.5. Other Applications

SHSs are increasingly leveraged for a wide range of innovative and interdisciplinary applications in both clinical and non-clinical settings. SHSs provide an ideal genomic environment for the stable integration of synthetic gene circuits. This is particularly valuable in engineering mammalian cells to perform complex tasks such as environmental sensing and synthesis of pharmaceuticals [189]. Targeting synthetic elements into SHSs ensures uniform expression across cell populations, reducing variability and positional effects that can compromise circuit function [189]. SHSs also offer strategic advantages for generating transgenic plants and animals with enhanced traits, disease resistance, or improved nutritional profiles, while minimizing the risks of insertional mutagenesis and transgene silencing [190,191]. In addition, they can serve as secure integration sites for biosensors and reporter genes designed to detect metabolic cues, track biological processes, or monitor environmental toxins and pathogens [192,193]. Stable sensor expression from SHSs ensures consistent functionality across cell generations, a feature essential for reproducibility and regulatory compliance [193,194]. Altogether, these emerging applications place SHSs as versatile platforms for safe and stable transgene expression. As more context-specific SHSs are identified, their utility in next-generation genomic engineering will continue to expand substantially.

6. Conclusions, Challenges, and Future Perspective

SHSs represent a pivotal advancement in the field of transgenesis, genetic engineering, and gene therapy. By providing reliable genomic locations for transgene integration with minimal disruption to native gene function or genomic integrity, SHSs have become invaluable tools for diverse applications from studying gene function and modeling human diseases to supporting drug discovery, producing biopharmaceuticals, and building genetic circuits for complex bioprocesses.
Despite their promises, the implementation of SHSs for a wider range of applications is hindered by several challenges. First, identifying and validating SHSs requires extensive genomic and epigenetic profiling to ensure they do not interfere with essential regulatory elements. This is further complicated by our incomplete understanding of the human genome and epigenome, compounded by the fact that epigenetic landscapes vary among individuals and can change in response to environmental factors. Second, epigenetic-mediated transgene silencing remains a major challenge in transgenesis and genetic engineering [16]. Given our limited understanding of human epigenetic regulation, it is difficult to predict or prevent silencing events of SHS-integrated transgenes, which have been reported in several cellular models, including iPSCs-derived cardiomyocytes, hepatocytes, and myeloid cells [100,101,195]. Third, the transgene targeting technologies, such as CRISPR/Cas9, can cause off-target effects with unintended consequences, also due to DNA double-strand breaks required for integration. Fourth, targeted gene integration into SHSs is inherently inefficient and is often outcompeted by faulty DNA repair, leading to insertions or deletions. Fifth, methods for delivering genome editing components together with donor templates remain inefficient, particularly for large inserts, and can induce cytotoxicity. Finally, clinical use of SHSs raises important ethical questions about consent, privacy, and possible social impacts. Especially, germline editing raises concerns over “designer babies” and the broader ethical consequences of altering the human germline.
Future studies should focus systematically on addressing these biological, technical, and ethical challenges while leveraging the constant advances in genome engineering. Integrating multi-omics data, including transcriptomic, epigenomic, and proteomic profiles, will improve the precision and robustness of SHS identification, ensuring the selection of loci that remain consistently active across different cell types and individuals. Additionally, the creation of synthetic SHSs, strategically inserted into well-characterized genomic regions, could be a valid strategy to avoid transgene silencing and mitigate unpredictable epigenetic effects, enabling more consistent and durable gene expression. Moreover, innovations in precision editing, such as high-fidelity Cas9 variants and prime editing, can mitigate off-target activity, while transient inhibition of non-homologous end joining could enhance homology-directed repair and thereby improve targeted integration efficiency. Advances in nanoparticle-based delivery systems may further reduce toxicity and facilitate the insertion of large genetic payloads in therapeutic contexts. Additionally, the increasing availability of population-scale genomic datasets could support the development of personalized SHS-insertion strategies. Tailoring the insertion to an individual’s genomic and epigenetic landscape can improve both safety and efficacy, particularly in patient-specific approaches such as iPSC-based regenerative therapies. Finally, ethical oversight must evolve in parallel with technical capabilities. Robust global guidelines and transparent public discourse are essential to ensure informed consent, protect genetic privacy, and promote equitable access while guarding against misuse. This is particularly important as germline editing becomes technically more feasible and socially contentious. In summary, the use of SHSs holds exceptional promise for the future of genetic engineering, functional genomics, and gene-based therapies. However, realizing their full potential will require overcoming considerable scientific, technological, and ethical barriers. With continued innovation, rigorous validation, and responsible governance, SHSs could become foundational elements of precise, safe, and effective genome engineering in both research and clinical settings.

Author Contributions

Conceptualization, A.A., V.C. and G.F.; writing—original draft preparation, A.A., D.D.M., G.N.I., A.C., A.S., A.B., L.P. and V.D.; writing—review and editing, A.A., D.D.M., G.N.I., A.C., A.S., A.B., L.P. and V.D.; supervision, V.C., L.P. and G.F.; funding acquisition, G.F. and L.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the European Union—NextGenerationEU—Project code CN00000041, “National Center for Gene Therapy and Drugs based on RNA Technology”—Spoke 1.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the authors used BioRender.com for the purposes of figure visualization. The authors have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Naidoo, N.; Pawitan, Y.; Soong, R.; Cooper, D.N.; Ku, C.S. Human genetics and genomics a decade after the release of the draft sequence of the human genome. Hum. Genom. 2011, 5, 577–622. [Google Scholar] [CrossRef]
  2. Marian, A.J. Sequencing your genome: What does it mean? Methodist DeBakey Cardiovasc. J. 2014, 10, 3–6. [Google Scholar] [CrossRef] [PubMed]
  3. Comfort, N. Genetics: We are the 98%. Nature 2015, 520, 615–616. [Google Scholar] [CrossRef]
  4. Fu, X.D. Non-coding RNA: A new frontier in regulatory biology. Natl. Sci. Rev. 2014, 1, 190–204. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, P.; Wu, W.; Chen, Q.; Chen, M. Non-coding RNAs and their integrated networks. J. Integr. Bioinform. 2019, 16, 20190027. [Google Scholar] [CrossRef] [PubMed]
  6. Riethoven, J.J.M. Regulatory regions in DNA: Promoters, enhancers, silencers, and insulators. Methods Mol. Biol. 2010, 674, 33–42. [Google Scholar]
  7. Jayavelu, N.D.; Jajodia, A.; Mishra, A.; Hawkins, R.D. Candidate silencer elements for the human and mouse genomes. Nat. Commun. 2020, 11, 1061. [Google Scholar] [CrossRef]
  8. Fan, H.; Chu, J.-Y. A brief review of short tandem repeat mutation. Genom. Proteom. Bioinform. 2007, 5, 7–14. [Google Scholar] [CrossRef]
  9. Chehelgerdi, M.; Chehelgerdi, M.; Khorramian-Ghahfarokhi, M.; Shafieizadeh, M.; Mahmoudi, E.; Eskandari, F.; Rashidi, M.; Arshi, A.; Farsani, M.A. Comprehensive review of CRISPR-based gene editing: Mechanisms, challenges, and applications in cancer therapy. Mol. Cancer 2024, 23, 9. [Google Scholar] [CrossRef]
  10. Milone, M.C.; O’Doherty, U. Clinical use of lentiviral vectors. Leukemia 2018, 32, 1529–1541. [Google Scholar] [CrossRef]
  11. Bire, S.; Dusserre, Y.; Bigot, Y.; Mermod, N. PiggyBac transposase and transposon derivatives for gene transfer targeting the ribosomal DNA loci of CHO cells. J. Biotechnol. 2021, 341, 103–112. [Google Scholar] [CrossRef]
  12. Ranzani, M.; Cesana, D.; Bartholomae, C.C.; Sanvito, F.; Pala, M.; Benedicenti, F.; Pierangela Gallina, P.; Sergi, S.L.; Merella, S.; Bulfone, A.; et al. Lentiviral vector-based insertional mutagenesis identifies genes associated with liver cancer. Nat. Methods 2013, 10, 155–161. [Google Scholar] [CrossRef]
  13. Loeb, K.R.; Hughes, B.T.; Fissel, B.M.; Osteen, N.J.; Knoblaugh, S.E.; Grim, J.E.; Drury, J.L.; Sarver, A.; Dupuy, J.A.; Clurman, E.B. Insertional mutagenesis using the Sleeping Beauty transposon system identifies drivers of erythroleukemia in mice. Sci. Rep. 2019, 9, 5488. [Google Scholar] [CrossRef]
  14. Hacein-Bey-Abina, S.; Von Kalle, C.; Schmidt, M.; McCormack, M.P.; Wulffraat, N.; Leboulch, P.; Lim, A.; Osborne, C.S.; Pawliuk, R.; Morillon, E.; et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003, 302, 415–441. [Google Scholar] [CrossRef]
  15. Groves, I.J.; Drane, E.L.A.; Michalski, M.; Monahan, J.M.; Scarpini, C.G.; Smith, S.P.; Stanley, M.A.; Parish, J.L.; McBride, A.A.; Doorbar, J.; et al. Short- and long-range cis interactions between integrated HPV genomes and cellular chromatin dysregulate host gene expression in early cervical carcinogenesis. PLoS Pathog. 2021, 17, e1009879. [Google Scholar] [CrossRef] [PubMed]
  16. Cabrera, A.; Edelstein, H.I.; Glykofrydis, F.; Love, K.S.; Palacios, S.; Tycko, J.; Zhang, M.; Lensch, S.; Shields, C.E.; Livingston, M.; et al. The sound of silence: Transgene silencing in mammalian cell engineering. Cell Syst. 2022, 13, 950–973. [Google Scholar] [CrossRef] [PubMed]
  17. Sakurai, K.; Shimoji, M.; Tahimic, C.G.T.; Aiba, K.; Kawase, E.; Hasegawa, K.; Amagai, Y.; Suemori, H.; Nakatsuji, N. Efficient integration of transgenes into a defined locus in human embryonic stem cells. Nucleic Acids Res. 2010, 38, e96. [Google Scholar] [CrossRef] [PubMed]
  18. Woo, H.J.; Kim, J.; Kim, S.M.; Kim, D.; Moon, J.Y.; Park, D.; Lee, E.; Choi, S.; Jeong, H.; Yang, K.; et al. Context-dependent genomic locus effects on antibody production in recombinant Chinese hamster ovary cells generated through random integration. Comput. Struct. Biotechnol. J. 2024, 23, 1654–1665. [Google Scholar] [CrossRef]
  19. Kimura, Y.; Shofuda, T.; Higuchi, Y.; Nagamori, I.; Oda, M.; Nakamori, M.; Onodera, M.; Kanematsu, D.; Yamamoto, A.; Katsuma, A.; et al. Human genomic safe harbors and the suicide gene-based safeguard system for iPSC-based cell therapy. Stem Cells Transl. Med. 2019, 8, 627–638. [Google Scholar] [CrossRef]
  20. Pavani, G.; Amendola, M. Targeted Gene Delivery: Where to Land. Front. Genome Edit 2021, 2, 609650. [Google Scholar] [CrossRef]
  21. Papapetrou, E.P.; Schambach, A. Gene Insertion Into Genomic Safe Harbors for Human Gene Therapy. Mol. Ther. 2016, 24, 678–684. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, X.; Yang, G.; Ji, P.; Liu, G.; Zhang, L. Identification of Site in the UTY Gene as Safe Harbor Locus on the Y Chromosome of Pig. Genes 2024, 15, 1005. [Google Scholar] [CrossRef] [PubMed]
  23. Shrestha, D.; Bag, A.; Wu, R.; Zhang, Y.; Tang, X.; Qi, Q.; Xing, J.; Cheng, Y. Genomics and epigenetics guided identification of tissue-specific genomic safe harbors. Genome Biol. 2022, 23, 199. [Google Scholar] [CrossRef] [PubMed]
  24. Kotin, R.M.; Linden, R.M.; Berns, K.I. Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. Embo J. 1992, 11, 5071–5078. [Google Scholar] [CrossRef]
  25. Merling, R.K.; Sweeney, C.L.; Chu, J.; Bodansky, A.; Choi, U.; Priel, D.L.; Kuhns, D.B.; Wang, H.; Vasilevsky, S.; De Ravin, S.; et al. An AAVS1-targeted minigene platform for correction of iPSCs from all five types of chronic granulomatous disease. Mol. Ther. 2015, 23, 147–157. [Google Scholar] [CrossRef]
  26. Oceguera-Yañez, F.; Kim, S.I.; Matsumoto, T.; Tan, G.W.; Xiang, L.; Hatani, T.; Mochizuki, M.; Bando, T.; Hatta, T.; Fukasawa, M.; et al. Engineering the AAVS1 locus for consistent and scalable transgene expression in human iPSCs and their differentiated derivatives. Methods 2016, 101, 43–55. [Google Scholar] [CrossRef]
  27. Irion, S.; Luche, H.; Gadue, P.; Fehling, H.J.; Kennedy, M.; Keller, G. Identification and targeting of the ROSA26 locus in human embryonic stem cells. Nat. Biotechnol. 2007, 25, 1477–1482. [Google Scholar] [CrossRef]
  28. Nyabi, O.; Naessens, M.; Haigh, K.; Gembarska, A.; Goossens, S.; Maetens, M.; De Clercq, S.; Drogat, B.; Haenebalcke, L.; Bartunkova, S.; et al. Efficient mouse transgenesis using Gateway-compatible ROSA26 locus targeting vectors and F1 hybrid ES cells. Nucleic Acids Res. 2009, 37, e55. [Google Scholar] [CrossRef]
  29. Wang, C.; Fang, S.; Chen, Y.; Tang, N.; Jiao, G.; Hu, Y.; Li, J.; Shan, Q.; Wang, X.; Feng, G.; et al. High-efficiency targeted transgene integration via primed micro-homologues. Cell Discov. 2023, 9, 69. [Google Scholar] [CrossRef]
  30. Knipping, F.; Osborn, M.J.; Petri, K.; Tolar, J.; Glimm, H.; von Kalle, C.; Pereira, L.; Deyell, R.J.; Wagner, J.E.; Woodard, K.; et al. Genome-wide specificity of highly efficient TALENs and CRISPR/Cas9 for T cell receptor modification. Mol. Ther. Methods Clin. Dev. 2017, 4, 213–224. [Google Scholar] [CrossRef]
  31. Li, Y.S.; Meng, R.R.; Chen, X.; Shang, C.L.; Li, H.B.; Zhang, T.J.; Long, H.Y.; Li, H.Q.; Wang, Y.J.; Wang, F.C.; et al. Generation of H11-albumin-rtTA transgenic mice: A tool for inducible gene expression in the liver. G3 2019, 9, 591–599. [Google Scholar] [CrossRef]
  32. Landau, D.J.; Brooks, E.D.; Perez-Pinera, P.; Amarasekara, H.; Mefferd, A.; Li, S.; Kaestner, K.H.; Porteus, M.H.; Wilson, M.H.; Yant, S.R.; et al. In vivo zinc finger nuclease-mediated targeted integration of a glucose-6-phosphatase transgene promotes survival in mice with glycogen storage disease type IA. Mol. Ther. 2016, 24, 697–706. [Google Scholar] [CrossRef]
  33. Pantazis, C.B.; Yang, A.; Lara, E.; McDonough, J.A.; Blauwendraat, C.; Peng, L.; Tang, J.; Liao, Z.; Sun, N.; Gros-Laux, I.; et al. A reference human induced pluripotent stem cell line for large-scale collaborative studies. Cell Stem Cell 2022, 29, 1685–1702. [Google Scholar] [CrossRef] [PubMed]
  34. Cerneckis, J.; Cai, H.; Shi, Y. Induced pluripotent stem cells (iPSCs): Molecular mechanisms of induction and applications. Signal Transduct. Target. Ther. 2024, 9, 112. [Google Scholar] [CrossRef] [PubMed]
  35. Deng, Y.; Ding, X.; Wu, Q.; Cai, Y.; Jin, J.; Xu, Q.; Zhang, S.; Li, H.; Chen, Y.; Wang, J.; et al. Construction of a stable expression HEK293T engineered cell line adapted to high-density suspension culture. Biochem. Eng. J. 2024, 207, 109334. [Google Scholar] [CrossRef]
  36. Aboul-Soud, M.A.M.; Alzahrani, A.J.; Mahmoud, A. Induced pluripotent stem cells (iPSCs)-roles in regenerative therapies, disease modelling and drug screening. Cells 2021, 10, 2319. [Google Scholar] [CrossRef]
  37. Lienert, F.; Lohmueller, J.J.; Garg, A.; Silver, P.A. Synthetic biology in mammalian cells: Next generation research tools and therapeutics. Nat. Rev. Mol. Cell Biol. 2014, 15, 95–107. [Google Scholar] [CrossRef] [PubMed]
  38. Cantos, C.; Francisco, P.; Trijatmiko, K.R.; Slamet-Loedin, I.; Chadha-Mohanty, P.K. Identification of “safe harbor” loci in indica rice genome by harnessing the property of zinc-finger nucleases to induce DNA damage and repair. Front. Plant Sci. 2014, 26, 302. [Google Scholar]
  39. Carroll, D. Genome Engineering with Targetable Nucleases. Annu. Rev. Biochem. 2014, 83, 409–439. [Google Scholar] [CrossRef]
  40. Wu, X.; Scott, D.A.; Kriz, A.J.; Chiu, A.C.; Hsu, P.D.; Dadon, D.B.; Cheng, A.W.; Trevino, A.E.; Konermann, S.; Chen, S.; et al. Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat. Biotechnol. 2014, 32, 670–676. [Google Scholar] [CrossRef]
  41. Zerbino, D.R.; Frankish, A.; Flicek, P. Progress, Challenges, and Surprises in Annotating the Human Genome. Annu. Rev. Genom. Hum. Genet. 2020, 21, 55–79. [Google Scholar] [CrossRef]
  42. Stuart, T.; Satija, R. Integrative single-cell analysis. Nat. Rev. Gen. 2019, 20, 257–272. [Google Scholar] [CrossRef] [PubMed]
  43. Quezada-Ramírez, M.A.; Loncar, S.; Campbell, M.A.; Parsi, K.M.; Gifford, R.J.; Kotin, R.M. Development of evolutionarily conserved viral integration sites as safe harbors for human gene therapy. iScience 2025, 28, 113910. [Google Scholar] [CrossRef] [PubMed]
  44. Odak, A.; Yuan, H.; Feucht, J.; Cantu, V.A.; Mansilla-Soto, J.; Kogel, F.; Eyquem, J.; Everett, J.; Bushman, F.D.; Leslie, C.S.; et al. Novel extragenic genomic safe harbors for precise therapeutic T-cell engineering. Blood 2023, 141, 2698–2712. [Google Scholar] [CrossRef]
  45. Papapetrou, E.P.; Lee, G.; Malani, N.; Setty, M.; Riviere, I.; Tirunagari, L.M.; Kadota, K.; Roth, S.L.; Giardina, P.; Viale, A.; et al. Genomic safe harbors permit high β-globin transgene expression in thalassemia induced pluripotent stem cells. Nat. Biotechnol. 2011, 29, 73–78. [Google Scholar] [CrossRef]
  46. Sadelain, M.; Papapetrou, E.P.; Bushman, F.D. Safe harbours for the integration of new DNA in the human genome. Nat. Rev. Cancer 2011, 12, 51–58. [Google Scholar] [CrossRef]
  47. Gilbertson, S.E.; Walter, H.C.; Gardner, K.; Wren, S.N.; Vahedi, G.; Weinmann, A.S. Topologically associating domains are disrupted by evolutionary genome rearrangements forming species-specific enhancer connections in mice and humans. Cell Rep. 2022, 39, 110769. [Google Scholar] [CrossRef]
  48. Lupiáñez, D.G.; Kraft, K.; Heinrich, V.; Krawitz, P.; Brancati, F.; Klopocki, E.; Horn, D.; Kayserili, H.; Opitz, J.M.; Laxova, R.; et al. Disruptions of topological chromatin domains cause pathogenic rewiring of gene-enhancer interactions. Cell 2015, 161, 1012–1025. [Google Scholar] [CrossRef]
  49. Miele, A.; Dekker, J. Long-range chromosomal interactions and gene regulation. Mol. Biosyst. 2008, 4, 1046–1057. [Google Scholar] [CrossRef]
  50. Tsagakis, I.; Douka, K.; Birds, I.; Aspden, J.L. Long non-coding RNAs in development and disease: Conservation to mechanisms. J. Pathol. 2020, 250, 480–495. [Google Scholar] [CrossRef] [PubMed]
  51. Aznauryan, E.; Yermanos, A.; Kinzina, E.; Devaux, A.; Kapetanovic, E.; Milanova, D.; Church, G.M.; Reddy, S.T. Discovery and validation of human genomic safe harbor sites for gene and cell therapies. Cell Rep. Methods 2022, 2, 100154. [Google Scholar] [CrossRef]
  52. Ciabrelli, F.; Cavalli, G. Chromatin-driven behavior of topologically associating domains. J. Mol. Biol. 2015, 427, 608–625. [Google Scholar] [CrossRef] [PubMed]
  53. Barral, A.; Déjardin, J. The chromatin signatures of enhancers and their dynamic regulation. Nucleus 2023, 14, 2160551. [Google Scholar] [CrossRef]
  54. Bressan, R.B.; Dewari, P.S.; Kalantzaki, M.; Gangoso, E.; Matjusaitis, M.; Garcia-Diaz, C.; Blin, C.; Grant, V.; Bulstrode, H.; Gogolok, S.; et al. Efficient CRISPR/Cas9-assisted gene targeting enables rapid and precise genetic manipulation of mammalian neural stem cells. Development 2017, 144, 635–648. [Google Scholar] [CrossRef] [PubMed]
  55. Schjeide, B.M.M.; Püschel, G.P. Determining on-target, off-target, and copy number status of transgenic events after CRISPR/Cas9-targeted AAVS1 safe-harbor modification of iPSCs using double-control quantitative copy number PCR. Curr. Protoc. 2023, 3, e635. [Google Scholar] [CrossRef] [PubMed]
  56. Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of genome editing technology in the targeted therapy of human diseases: Mechanisms, advances and prospects. Signal Transduct Target Ther. 2020, 5, 1. [Google Scholar] [CrossRef]
  57. Hermantara, R.; Richmond, L.; Taqi, A.F.; Chilaka, S.; Jeantet, V.; Guerrini, I.; West, K.; West, A. Improving CRISPR–Cas9-directed faithful transgene integration outcomes by reducing unwanted random DNA integration. J. Biomed. Sci. 2024, 31, 32. [Google Scholar] [CrossRef] [PubMed]
  58. Guo, C.; Ma, X.; Gao, F.; Guo, Y. Off-target effects in CRISPR/Cas9 gene editing. Front. Bioeng. Biotechnol. 2023, 11. [Google Scholar] [CrossRef]
  59. Mazan-Mamczarz, K.; Ha, J.; De, S.; Sen, P. Single-Cell Analysis of the Transcriptome and Epigenome. Methods Mol. Biol. 2022, 2399, 21–60. [Google Scholar]
  60. Mudge, J.M.; Carbonell-Sala, S.; Diekhans, M.; Martinez, J.G.; Hunt, T.; Jungreis, I.; Loveland, J.E.; Arnan, C.; Barnes, I.; Bennett, R.; et al. GENCODE 2025: Reference gene annotation for human and mouse. Nucleic Acids Res. 2024, 53, D966–D975. [Google Scholar] [CrossRef]
  61. Guigó, R. Genome annotation: From human genetics to biodiversity genomics. Cell Genom. 2023, 3, 100375. [Google Scholar] [CrossRef]
  62. Knowles, D.A.; Davis, J.R.; Edgington, H.; Raj, A.; Favé, M.J.; Zhu, X.; Potash, J.B.; Weissman, M.M.; Shi, J.; Levinson, D.F.; et al. Allele-specific expression reveals interactions between genetic variation and environment. Nat. Methods 2017, 14, 699–702. [Google Scholar] [CrossRef] [PubMed]
  63. Heinz, S.; Romanoski, C.E.; Benner, C.; Glass, C.K. The selection and function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 2015, 16, 144–154. [Google Scholar] [CrossRef]
  64. Breton, C.V.; Landon, R.; Kahn, L.G.; Enlow, M.B.; Peterson, A.K.; Bastain, T.; Braun, J.; Comstock, S.S.; Duarte, C.S.; Hipwell, A. Exploring the evidence for epigenetic regulation of environmental influences on child health across generations. Commun. Biol. 2021, 4, 769. [Google Scholar] [CrossRef]
  65. Capp, J.P. Interplay between genetic, epigenetic, and gene expression variability: Considering complexity in evolvability. Evol. Appl. 2021, 14, 893–901. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, B.; Ren, C.; Ouyang, Z.; Xu, J.; Xu, K.; Li, Y.; Guo, H.; Bai, X.; Tian, M.; Xu, X.; et al. Stratifying TAD boundaries pinpoints focal genomic regions of regulation, damage, and repair. Brief. Bioinform. 2024, 25, bbae306. [Google Scholar] [CrossRef] [PubMed]
  67. Rajderkar, S.; Barozzi, I.; Zhu, Y.; Hu, R.; Zhang, Y.; Li, B.; Alcaina Caro, A.; Fukuda-Yuzawa, Y.; Kelman, G.; Akeza, A.; et al. Topologically associating domain boundaries are required for normal genome function. Commun. Biol. 2023, 6, 435. [Google Scholar] [CrossRef]
  68. Boob, A.G.; Zhu, Z.; Intasian, P.; Jain, M.; Petrov, V.A.; Lane, S.T.; Tan, S.-I.; Xun, G.; Zhao, H. CRISPR-COPIES: An in silico platform for discovery of neutral integration sites for CRISPR/Cas-facilitated gene integration. Nucleic Acids Res. 2024, 52, e30. [Google Scholar] [CrossRef]
  69. Autio, M.I.; Motakis, E.; Perrin, A.; Bin Amin, T.; Tiang, Z.; Do, D.V.; Wang, J.; Tan, J.K.M.; Ding, S.S.L.; Tan, W.X.; et al. Computationally defined and in vitro validated putative genomic safe harbour loci for transgene expression in human cells. eLife 2024, 13, e79592. [Google Scholar] [CrossRef]
  70. Biffi, A.; Bartolomae, C.C.; Cesana, D.; Cartier, N.; Aubourg, P.; Ranzani, M.; Calabria, A.; Canale, S.; Leimer, K.; Lattanzi, A.; et al. Lentiviral vector common integration sites in preclinical models and a clinical trial reflect a benign integration bias and not oncogenic selection. Blood 2011, 117, 5332–5339. [Google Scholar] [CrossRef]
  71. Monse, H.; Laufs, S.; Kuate, S.; Zeller, W.J.; Fruehauf, S.; Uberla, K. Viral determinants of integration site preferences of simian immunodeficiency virus-based vectors. J. Virol. 2006, 80, 8145–8150. [Google Scholar] [CrossRef]
  72. Poletti, V.; Mavilio, F. Interactions between Retroviruses and the Host Cell Genome. Mol. Ther. Methods Clin. Dev. 2017, 8, 31–41. [Google Scholar] [CrossRef]
  73. Costa, M.; Dottori, M.; Ng, E.; Hawes, S.M.; Sourris, K.; Jamshidi, P.; Pera, M.F.; Elefanty, A.G.; Stanley, E.G. The hESC line Envy expresses high levels of GFP in all differentiated progeny. Nat. Methods 2005, 2, 259–260. [Google Scholar] [CrossRef]
  74. Figueroa-Bossi, N.; Balbontín, R.; Bossi, L. Mapping transposon insertion sites by inverse polymerase chain reaction and Sanger sequencing. Cold Spring Harb. Protoc. 2024, 5, 108197. [Google Scholar]
  75. Kim, H.S.; Lee, H.; Bae, T.; Park, S.-H.; Kim, Y.J.; Lee, S.; Lee, S.; Park, J.H.; Bae, S.; Hur, J.K. CReVIS-Seq: A highly accurate and multiplexable method for genome-wide mapping of lentiviral integration sites. Mol. Ther. Methods Clin. Dev. 2021, 20, 193–204. [Google Scholar] [CrossRef]
  76. Liu, Y.; Thyagarajan, B.; Lakshmipathy, U.; Xue, H.; Lieu, P.; Fontes, A.; MacArthur, C.C.; Scheyhing, K.; Rao, M.S.; Chesnut, J.D. Generation of platform human embryonic stem cell lines that allow efficient targeting at a predetermined genomic location. Stem Cells Dev. 2009, 18, 1459–1472. [Google Scholar] [CrossRef] [PubMed]
  77. Ustek, D.; Sirma, S.; Gumus, E.; Arikan, M.; Cakiris, A.; Abaci, N.; Mathew, J.; Emrence, Z.; Azakli, H.; Cosan, F.; et al. A genome-wide analysis of lentivector integration sites using targeted sequence capture and next-generation sequencing technology. Infect. Genet. Evol. 2012, 12, 1349–1354. [Google Scholar] [CrossRef]
  78. Kelly, J.J.; Saee-Marand, M.; Nyström, N.N.; Evans, M.M.; Chen, Y.; Martinez, F.M.; Hamilton, A.M.; Ronald, J.A. Safe harbor-targeted CRISPR-Cas9 homology-independent targeted integration for multimodality reporter gene-based cell tracking. Sci. Adv. 2021, 7, eabc3791. [Google Scholar] [CrossRef]
  79. Liao, H.; Wu, J.; VanDusen, N.J.; Li, Y.; Zheng, Y. CRISPR-Cas9-mediated homology-directed repair for precise gene editing. Mol. Ther. Nucleic Acids 2024, 35, 102057. [Google Scholar] [CrossRef] [PubMed]
  80. Pellenz, S.; Phelps, M.; Tang, W.; Hovde, B.T.; Sinit, R.B.; Fu, W.; Li, H.; Chen, E.; Monnat, R.J. New human chromosomal sites with “safe harbor” potential for targeted transgene insertion. Hum. Gene Ther. 2019, 30, 814–828. [Google Scholar] [CrossRef]
  81. Chaudhari, N.; Rickard, A.M.; Roy, S.; Dröge, P.; Makhija, H. A non-viral genome editing platform for site-specific insertion of large transgenes. Stem Cell Res. Ther. 2020, 11, 380. [Google Scholar] [CrossRef]
  82. Samulski, R.J.; Zhu, X.; Xiao, X.; Brook, J.D.; Housman, D.E.; Epstein, N.; Hunter, L.A.; Laughlin, C.A. Targeted integration of adeno-associated virus (AAV) into human chromosome 19. EMBO J. 1991, 10, 3941–3950. [Google Scholar] [CrossRef]
  83. Kotin, R.M.; Menninger, J.C.; Ward, D.C.; Berns, K.I. Mapping and direct visualization of a region-specific viral DNA integration site on chromosome 19q13–qter. Genomics 1991, 10, 831–834. [Google Scholar] [CrossRef]
  84. Kotin, R.M.; Siniscalco, M.; Samulski, R.J.; Zhu, X.D.; Hunter, L.; Laughlin, C.A.; McLaughlin, S.; Muzyczka, N.; Berns, K.I. Site-specific integration by adeno-associated virus. Proc. Natl. Acad. Sci. USA 1990, 87, 2211–2215. [Google Scholar] [CrossRef] [PubMed]
  85. Musayev, F.N.; Zarate-Perez, F.; Bishop, C.; Burgner, J.W., II; Escalante, C.R. Structural insights into the assembly of the adeno-associated virus type 2 Rep68 protein on the integration site AAVS1. J. Biol. Chem. 2015, 290, 27487–27499. [Google Scholar] [CrossRef]
  86. McCarty, D.M.; Young, S.M.; Samulski, R.J. Integration of adeno-associated virus (AAV) and recombinant AAV vectors. Annu Rev Genet. 2004, 38, 819–845. [Google Scholar] [CrossRef] [PubMed]
  87. Hong, S.; Lee, J.; Kim, Y.; Kim, E.; Shin, K. AAVS1-targeted, stable expression of ChR2 in human brain organoids for consistent optogenetic control. Bioeng. Transl. Med. 2024, 9, e10690. [Google Scholar] [CrossRef]
  88. Smith, J.R.; Maguire, S.; Davis, L.A.; Alexander, M.; Yang, F.; Chandran, S.; ffrench-Constant, C.; Pedersen, R.A. Robust, persistent transgene expression in human embryonic stem cells is achieved with AAVS1-targeted integration. Stem Cells 2008, 26, 496–504. [Google Scholar] [CrossRef]
  89. Hockemeyer, D.; Soldner, F.; Beard, C.; Gao, Q.; Mitalipova, M.; DeKelver, R.C.; Katibah, G.E.; Amora, R.; Boydston, E.A.; Zeitler, B.; et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat. Biotechnol. 2009, 27, 851–857. [Google Scholar] [CrossRef]
  90. DeKelver, R.C.; Choi, V.M.; Moehle, E.A.; Paschon, D.E.; Hockemeyer, D.; Meijsing, S.H.; Sancak, Y.; Cui, X.; Steine, E.J.; Miller, J.C.; et al. Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human genome. Genome Res. 2010, 20, 1133–1142. [Google Scholar] [CrossRef]
  91. Li, S.J.; Luo, Y.; Zhang, L.M.; Yang, W.; Zhang, G.G. Targeted introduction and effective expression of hFIX at the AAVS1 locus in mesenchymal stem cells. Mol. Med. Rep. 2017, 15, 1313–1318. [Google Scholar] [CrossRef]
  92. Kajimura, Y.; Dong, S.; Tessari, A.; Orlacchio, A.; Thoms, A.; Cufaro, M.C.; Fries, B.; Zhang, L.; Mancini, R.; Amann, J.; et al. An in vivo “turning model” reveals new RanBP9 interactions in lung macrophages. Cell Death Discov. 2025, 11, 171. [Google Scholar] [CrossRef] [PubMed]
  93. Li, C.; Mishra, A.S.; Gil, S.; Wang, M.; Georgakopoulou, A.; Papayannopoulou, T.; Hawkins, R.D.; Lieber, A. Targeted Integration and High-Level Transgene Expression in AAVS1 Transgenic Mice after In Vivo HSC Transduction with HDAd5/35++ Vectors. Mol. Ther. 2019, 27, 2195–2212. [Google Scholar] [CrossRef]
  94. Ding, Y.; Wang, L.; Ji, W.; Chen, Z.; Wang, D.; Chen, C.; Li, X.; Zhang, Y.; Liu, J.; Sun, H.; et al. Generation of a human induced pluripotent stem cell line with Cas9 driven by Tet-on operator via AAVS1 safe harbor gene editing. Stem Cell Res. 2020, 49, 102064. [Google Scholar] [CrossRef]
  95. Zhang, K.; Wan, P.; Wang, L.; Wang, Z.; Tan, F.; Li, J.; Ma, X.; Cen, J.; Yuan, X.; Liu, Y.; et al. Efficient expansion and CRISPR-Cas9-mediated gene correction of patient-derived hepatocytes for treatment of inherited liver diseases. Cell Stem Cell 2024, 31, 1187–1202.e8. [Google Scholar] [CrossRef] [PubMed]
  96. Nadtochy, J.A.; Medvedev, S.P.; Grigor’eva, E.V.; Pavlova, S.V.; Minina, J.M.; Chechushkov, A.V.; Malakhova, A.A.; Kovalenko, L.V.; Zakian, S.M. Transgenic iPSC lines with genetically encoded MitoTimer to study mitochondrial biogenesis in dopaminergic neurons with tauopathy. Biomedicines 2025, 13, 550. [Google Scholar] [CrossRef]
  97. Orlacchio, A.; Kajimura, Y.; Rizzotto, L.; Tessari, A.; Soliman, S.H.A.; Visone, R.; Zhang, L.; Fries, B.; Tessarollo, L.; Amann, J.; et al. RANBP9 and RANBP10 cooperate in regulating non-small cell lung cancer proliferation. J. Exp. Clin. Cancer Res. 2025, 44, 259. [Google Scholar] [CrossRef] [PubMed]
  98. Karbassi, E.; Padgett, R.; Bertero, A.; Reinecke, H.; Klaiman, J.M.; Yang, X.; Hauschka, S.D.; Murry, C.E. Targeted CRISPR activation is functional in engineered human pluripotent stem cells but undergoes silencing after differentiation into cardiomyocytes and endothelium. Cell Mol. Life Sci. 2024, 81, 95. [Google Scholar] [CrossRef]
  99. Bhagwan, J.R.; Collins, E.; Mosqueira, D.; Bakar, M.; Johnson, B.B.; Thompson, A.; James, G.W.S.; Denning, C. Variable expression and silencing of CRISPR-Cas9 targeted transgenes identifies the AAVS1 locus as not an entirely safe harbour. F1000Res 2019, 8, 1234. [Google Scholar] [CrossRef]
  100. Klatt, D.; Cheng, E.; Hoffmann, D.; Santilli, G.; Thrasher, A.J.; Brendel, C.; Galla, M.; Kuehle, J.; Schneider, S.; Müller, J.; et al. Differential transgene silencing of myeloid-specific promoters in the AAVS1 safe harbor locus of induced pluripotent stem cell-derived myeloid cells. Hum. Gene Ther. 2020, 31, 199–210. [Google Scholar] [CrossRef]
  101. Ordovás, L.; Boon, R.; Pistoni, M.; Chen, Y.; Wolfs, E.; Guo, W.; Sambathkumar, R.; Bobis-Wozowicz, S.; Helsen, N.; Vanhove, J.; et al. Efficient recombinase-mediated cassette exchange in hPSCs to study the hepatocyte lineage reveals AAVS1 locus-mediated transgene inhibition. Stem Cell Rep. 2015, 5, 918–931. [Google Scholar] [CrossRef]
  102. Inderbitzin, A.; Loosli, T.; Kouyos, R.D.; Metzner, K.J. Quantification of transgene expression in GSH AAVS1 with a novel CRISPR/Cas9-based approach reveals high transcriptional variation. Mol. Ther. Methods Clin. Dev. 2022, 26, 107–118. [Google Scholar] [CrossRef]
  103. Mizutani, T.; Li, R.; Haga, H.; Kawabata, K. Transgene integration into the human AAVS1 locus enhances myosin II-dependent contractile force by reducing expression of myosin binding subunit 85. Biochem. Biophys. Res. Commun. 2015, 465, 270–274. [Google Scholar] [CrossRef]
  104. Cerbini, T.; Funahashi, R.; Luo, Y.; Liu, C.; Park, K.; Rao, M.; Zhang, Y.; Chen, J.; Li, H.; Wang, Q.; et al. Transcription activator-like effector nuclease (TALEN)-mediated CLYBL targeting enables enhanced transgene expression and one-step generation of dual reporter human induced pluripotent stem cell (iPSC) and neural stem cell (NSC) lines. PLoS ONE 2015, 10, e0139844. [Google Scholar] [CrossRef] [PubMed]
  105. Piovan, C.; Amari, F.; Lovat, F.; Chen, Q.; Coppola, V. Generation of mouse lines conditionally over-expressing microRNA using the Rosa26-Lox-Stop-Lox system. Methods Mol. Biol. 2014, 1215, 203–224. [Google Scholar]
  106. Kong, Q.; Hai, T.; Ma, J.; Huang, T.; Jiang, D.; Xie, B.; Liu, H.; Zhang, Y.; Li, F.; Wang, X.; et al. Rosa26 locus supports tissue-specific promoter driving transgene expression specifically in pig. PLoS ONE 2014, 9, e102551. [Google Scholar] [CrossRef]
  107. Yang, W.Q.; Xiong, Q.P.; Ge, J.Y.; Li, H.; Zhu, W.Y.; Nie, Y.; Zhang, L.; Chen, X.; Zhao, Y.; Liu, J.; et al. THUMPD3-TRMT112 is a m2G methyltransferase working on a broad range of tRNA substrates. Nucleic Acids Res. 2021, 49, 11900–11919. [Google Scholar] [CrossRef]
  108. Bertero, A.; Pawlowski, M.; Ortmann, D.; Snijders, K.; Yiangou, L.; Cardoso de Brito, M.; Garcia, M.; Khadayate, S.; Ng, S.Y.; Kilpinen, H.; et al. Optimized inducible shRNA and CRISPR/Cas9 platforms for in vitro studies of human development using hPSCs. Development 2016, 143, 4405–4418. [Google Scholar] [CrossRef]
  109. Speicher, A.M.; Korn, L.; Csatári, J.; Gonzalez-Cano, L.; Heming, M.; Thomas, C.; Reinwald, S.; Haenel, F.; Lyu, X.; Mokhtari, Z.; et al. Deterministic programming of human pluripotent stem cells into microglia facilitates studying their role in health and disease. Proc. Natl. Acad. Sci. USA 2022, 119, e2111836119. [Google Scholar] [CrossRef] [PubMed]
  110. Suzuki, R.; Kanemaki, M.T.; Suzuki, T.; Yoshioka, K. Overexpression of JNK-associated leucine zipper protein induces chromosomal instability through interaction with dynein light intermediate chain 1. Genes Cells 2024, 29, 39–51. [Google Scholar] [CrossRef]
  111. Shi, H.; Li, L.; Mu, S.; Gou, S.; Liu, X.; Chen, F.; Zhao, Y.; Wang, J.; Huang, W.; Tan, X.; et al. Exonuclease editor promotes precision of gene editing in mammalian cells. BMC Biol. 2024, 22, 119. [Google Scholar] [CrossRef] [PubMed]
  112. Goshayeshi, L.; Yousefi, T.S.; Dehdilani, N.; Nasiri, M.; Ghahramani Seno, M.M.; Dehghani, H. CRISPR/dCas9-mediated transposition with specificity and efficiency of site-directed genomic insertions. FASEB J. 2021, 35, e21896. [Google Scholar] [CrossRef]
  113. Zhu, F.; Gamboa, M.; Farruggio, A.P.; Hippenmeyer, S.; Tasic, B.; Schüle, B.; Chen, X.; Wernig, M.; Jaenisch, R.; Hockemeyer, D.; et al. DICE, an efficient system for iterative genomic editing in human pluripotent stem cells. Nucleic Acids Res. 2014, 42, e34. [Google Scholar] [CrossRef] [PubMed]
  114. Park, C.Y.; Sung, J.J.; Cho, S.R.; Kim, J.; Kim, D.W. Universal correction of blood coagulation factor VIII in patient-derived induced pluripotent stem cells using CRISPR/Cas9. Stem Cell Rep. 2019, 12, 1242–1249. [Google Scholar] [CrossRef]
  115. Pryzhkova, M.V.; Xu, M.J.; Jordan, P.W. Adaptation of the AID system for stem cell and transgenic mouse research. Stem Cell Res. 2020, 49, 102048. [Google Scholar] [CrossRef]
  116. Barmania, F.; Pepper, M.S. C-C chemokine receptor type five (CCR5): An emerging target for the control of HIV infection. Appl. Transl. Genom. 2013, 2, 3–16. [Google Scholar] [CrossRef]
  117. Liu, R.; Paxton, W.A.; Choe, S.; Ceradini, D.; Martin, S.R.; Horuk, R.; MacDonald, M.; Stuhlmann, H.; Koup, R.A.; Landau, N.R.; et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 1996, 86, 367–377. [Google Scholar] [CrossRef]
  118. Dragic, T.; Litwin, V.; Allaway, G.P.; Martin, S.R.; Huang, Y.; Nagashima, K.A.; Cayanan, C.; Maddon, P.J.; Koup, R.A.; Moore, J.P.; et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 1996, 381, 667–673. [Google Scholar] [CrossRef]
  119. Zamarchi, R.; Indraccolo, S.; Minuzzo, S.; Coppola, V.; Gringeri, A.; Santagostino, E.; Masiello, L.; Volpe, A.; Marcello, A.; Rizzi, M.; et al. Frequency of a mutated CCR-5 allele (delta32) among Italian healthy donors and individuals at risk of parenteral HIV infection. AIDS Res. Hum. Retroviruses 1999, 15, 337–344. [Google Scholar] [CrossRef]
  120. Kang, H.; Minder, P.; Park, M.A.; Mesquitta, W.T.; Torbett, B.E.; Slukvin, I.I. CCR5 disruption in induced pluripotent stem cells using CRISPR/Cas9 provides selective resistance of immune cells to CCR5-tropic HIV-1 virus. Mol. Ther. Nucleic Acids 2015, 4, e42. [Google Scholar] [CrossRef] [PubMed]
  121. Chen, G.; Jin, H.; Yu, Z.; Liu, Y.; Li, Z.; Navarengom, K.; Zhang, H.; Wang, L.; Li, J.; Zhao, X.; et al. Generation of human induced pluripotent stem cells from individuals with a homozygous CCR5Δ32 mutation. Stem Cell Res. 2019, 38, 101485. [Google Scholar] [CrossRef]
  122. Lombardo, A.; Genovese, P.; Beausejour, C.M.; Colleoni, S.; Lee, Y.L.; Kim, K.A.; Ando, D.; Urnov, F.D.; Galli, C.; Gregory, P.D.; et al. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nat. Biotechnol. 2007, 25, 1298–1306. [Google Scholar] [CrossRef]
  123. Rothemejer, F.H.; Lauritsen, N.P.; Juhl, A.K.; Schleimann, M.H.; König, S.; Søgaard, O.S.; Pedersen, L.; Jørgensen, S.; Nielsen, C.; Mikkelsen, S.; et al. Development of HIV-resistant CAR T cells by CRISPR/Cas-mediated CAR integration into the CCR5 locus. Viruses 2023, 15, 202. [Google Scholar] [CrossRef]
  124. Shin, S.; Kim, S.H.; Shin, S.W.; Grav, L.M.; Pedersen, L.E.; Lee, J.S.; Kim, D.; Lee, H.; Choi, Y.; Park, S.; et al. Comprehensive analysis of genomic safe harbors as target sites for stable expression of the heterologous gene in HEK293 cells. ACS Synth. Biol. 2020, 9, 1263–1269. [Google Scholar] [CrossRef]
  125. Lombardo, A.; Cesana, D.; Genovese, P.; Di Stefano, B.; Provasi, E.; Colombo, D.F.; Neri, S.; Magnani, Z.; Cathomen, T.; Thrasher, A.J. Site-specific integration and tailoring of cassette design for sustainable gene transfer. Nat. Methods 2011, 8, 861–869. [Google Scholar] [CrossRef]
  126. Lim, J.K.; Glass, W.G.; McDermott, D.H.; Murphy, P.M. CCR5: No longer a “good for nothing” gene—Chemokine control of West Nile virus infection. Trends Immunol. 2006, 27, 308–312. [Google Scholar] [CrossRef]
  127. Glass, W.G.; McDermott, D.H.; Lim, J.K.; Lekhong, S.; Yu, S.F.; Frank, W.A.; Pape, J.; Cheshier, R.C.; Murphy, P.M. CCR5 deficiency increases risk of symptomatic West Nile virus infection. J. Exp. Med. 2006, 203, 35–40. [Google Scholar] [CrossRef]
  128. Thyagarajan, B.; Liu, Y.; Shin, S.; Lakshmipathy, U.; Scheyhing, K.; Xue, H.; Du, Y.; Fu, W.; Noggle, S.; Verfaillie, C.M.; et al. Creation of engineered human embryonic stem cell lines using phiC31 integrase. Stem Cells 2008, 26, 119–126. [Google Scholar] [CrossRef]
  129. Strittmatter, L.; Li, Y.; Nakatsuka, N.J.; Calvo, S.E.; Grabarek, Z.; Mootha, V.K. CLYBL is a polymorphic human enzyme with malate synthase and β-methylmalate synthase activity. Hum. Mol. Genet. 2014, 23, 2313–2323. [Google Scholar] [CrossRef]
  130. Shen, H.; Campanello, G.C.; Flicker, D.; Grabarek, Z.; Hu, J.; Luo, C.; Banerjee, R.; Mootha, V.K. The human knockout gene CLYBL connects itaconate to vitamin B12. Cell 2017, 171, 771–782.e11. [Google Scholar] [CrossRef]
  131. Sivakumar, S.; Wang, Y.; Goetsch, S.C.; Pandit, V.; Wang, L.; Zhao, H.; Sundarrajan, A.; Armendariz, D.; Takeuchi, C.; Nzima, M.; et al. Benchmarking and optimizing Perturb-seq in differentiating human pluripotent stem cells. Stem Cell Rep. 2025, 20, 102713. [Google Scholar] [CrossRef]
  132. Miellet, S.; St Clair-Glover, M.; Maddock, M.; Dottori, M. Generation of a gene-edited H9 embryonic stem cell line carrying a DOX-inducible NGN2 expression cassette in the CLYBL locus. Stem Cell Res. 2024, 75, 103312. [Google Scholar] [CrossRef]
  133. Pei, Y.; Sierra, G.; Sivapatham, R.; Swistowski, A.; Rao, M.S.; Zeng, X. A platform for rapid generation of single and multiplexed reporters in human iPSC lines. Sci. Rep. 2015, 5, 9205. [Google Scholar] [CrossRef] [PubMed]
  134. Dräger, N.M.; Sattler, S.M.; Huang, C.T.L.; Teter, O.M.; Leng, K.; Hashemi, S.H.; Hong, J.; Aviles, G.; Clelland, C.D.; Zhan, L.; et al. A CRISPRi/a platform in human iPSC-derived microglia uncovers regulators of disease states. Nat. Neurosci. 2022, 25, 1149–1162. [Google Scholar] [CrossRef]
  135. Wang, Q.; Liu, J.; Janssen, J.M.; Gonçalves, M.A.F.V. Precise homology-directed installation of large genomic edits in human cells with cleaving and nicking high-specificity Cas9 variants. Nucleic Acids Res. 2023, 51, 3465–3484. [Google Scholar] [CrossRef]
  136. Blanch-Asensio, A.; van der Vaart, B.; Vinagre, M.; Groen, E.; Arendzen, C.; Freund, C.; Geijsen, N.; Mummery, C.L.; Davis, R.P. Generation of AAVS1 and CLYBL STRAIGHT-IN v2 acceptor human iPSC lines for integrating DNA payloads. Stem Cell Res. 2023, 66, 102991. [Google Scholar] [CrossRef]
  137. Ulge, U.Y.; Baker, D.A.; Monnat, R.J., Jr. Comprehensive computational design of mCreI homing endonuclease cleavage specificity for genome engineering. Nucleic Acids Res. 2011, 39, 4330–4339. [Google Scholar] [CrossRef] [PubMed]
  138. Li, H.; Monnat, R.J., Jr. Homing endonuclease target site specificity defined by sequential enrichment and next-generation sequencing of highly complex target site libraries. In Methods in Molecular Biology; Smith, J., Ed.; Homing Endonucleases; Humana Press: Totowa, NJ, USA, 2014; Volume 968, pp. 123–134. [Google Scholar]
  139. Pellenz, S.; Monnat, R.J., Jr. Identification and analysis of genomic homing endonuclease target sites. In Methods in Molecular Biology; Smith, J., Ed.; Homing Endonucleases; Humana Press: Totowa, NJ, USA, 2014; Volume 968, pp. 145–158. [Google Scholar]
  140. Hafez, M.; Hausner, G. Homing endonucleases: DNA scissors on a mission. Genome 2012, 55, 553–569. [Google Scholar] [CrossRef]
  141. Vlassis, A.; Jensen, T.L.; Mohr, M.; Jedrzejczyk, D.J.; Meng, X.; Kovacs, G.; Kaur, S.; Patel, S.; Li, H.; Zhang, Y.; et al. CRISPR-Cas12a-integrated transgenes in genomic safe harbors retain high expression in human hematopoietic iPSC-derived lineages and primary cells. iScience 2023, 26, 105315. [Google Scholar] [CrossRef] [PubMed]
  142. Yanagi, T.; Phen, S.F.; Ayala, J.; Aydin, D.E.; Jaramillo, S.; Truong, D.M. Termination sequence between an inducible promoter and ubiquitous chromatin opening element (UCOE) reduces gene expression leakage and silencing. J. Biol. Eng. 2025, 19, 29. [Google Scholar] [CrossRef]
  143. Yang, Y.; Dashi, A.; Soong, P.L.; Lin, K.H.; Tan, W.L.W.; Pan, B.; Lim, C.Y.; Chen, J.; Lee, H.Y.; Ho, D.K.; et al. Long noncoding RNA VENTHEART is required for ventricular cardiomyocyte specification and function. J. Mol. Cell. Cardiol. 2024, 197, 90–102. [Google Scholar] [CrossRef]
  144. Sonawane, A.R.; Platig, J.; Fagny, M.; Chen, C.-Y.; Paulson, J.N.; Lopes-Ramos, C.M.; DeMeo, D.L.; Quackenbush, J.; Glass, K.; Kuijjer, M.L.; et al. Understanding Tissue-Specific Gene Regulation. Cell Rep. 2017, 21, 1077–1088. [Google Scholar] [CrossRef]
  145. Balmas, E.; Sozza, F.; Bottini, S.; Ratto, M.L.; Savorè, G.; Becca, S.; Rimondini, R.; Bellin, M.; Tedesco, F.; Testa, G.; et al. Manipulating and studying gene function in human pluripotent stem cell models. FEBS Lett. 2023, 597, 2250–2287. [Google Scholar] [CrossRef]
  146. Zhang, F.; Meier, A.B.; Poch, C.M.; Tian, Q.; Engelhardt, S.; Sinnecker, D.; Duffy, J.; Knott, R.; Iffland, L.; Wilson, C. High-throughput optical action potential recordings in hiPSC-derived cardiomyocytes with a genetically encoded voltage indicator in the AAVS1 locus. Front. Cell Dev. Biol. 2022, 10, 849050. [Google Scholar] [CrossRef]
  147. Dost, A.F.M.; Moye, A.L.; Vedaie, M.; Tran, L.M.; Fung, E.; Heinze, D.; Xu, R.; Snyder, E.L.; Rawls, K.; Li, W. Organoids Model Transcriptional Hallmarks of Oncogenic KRAS Activation in Lung Epithelial Progenitor Cells. Cell Stem Cell 2020, 27, 663–678.e8. [Google Scholar] [CrossRef]
  148. Kim, H.; Selvaraj, S.; Kiley, J.; Azzag, K.; Garay, B.I.; Perlingeiro, R.C.R. Genomic Safe Harbor Expression of PAX7 for the Generation of Engraftable Myogenic Progenitors. Stem Cell Rep. 2021, 16, 10–19. [Google Scholar] [CrossRef] [PubMed]
  149. Harley, P.; Kerins, C.; Gatt, A.; Neves, G.; Riccio, F.; Machado, C.B.; Cheesbrough, A.; R’Bibo, L.; Burrone, J.; Lieberam, I. Aberrant axon initial segment plasticity and intrinsic excitability of ALS hiPSC motor neurons. Cell Rep. 2023, 42, 113509. [Google Scholar] [CrossRef]
  150. Schmid, B.; Holst, B.; Poulsen, U.; Jørring, I.; Clausen, C.; Rasmussen, M.; Sørensen, K.; Nielsen, A.; Petersen, L.; Hansen, T.; et al. Generation of two gene edited iPSC-lines carrying a DOX-inducible NGN2 expression cassette with and without GFP in the AAVS1 locus. Stem Cell Res. 2021, 52, 102240. [Google Scholar] [CrossRef] [PubMed]
  151. Shan, X.; Cramer, A.L.; Jeong, C.G. Protocol for generating NGN2 iPSC lines and large-scale human neuron production. STAR Protoc. 2025, 6, 27. [Google Scholar] [CrossRef]
  152. Shan, X.; Zhang, A.; Rezzonico, M.G.; Tsai, M.-C.; Sanchez-Priego, C.; Zhang, Y.; Chen, M.B.; Choi, M.; Andrade López, J.M.; Phu, L.; et al. Fully defined NGN2 neuron protocol reveals diverse signatures of neuronal maturation. Cell Rep. Methods 2024, 4, 100858. [Google Scholar] [CrossRef]
  153. Gu, J.; Rollo, B.; Sumer, H.; Cromer, B. Targeting the AAVS1 site by CRISPR/Cas9 with an inducible transgene cassette for the neuronal differentiation of human pluripotent stem cells. In Applications of Genome Modulation and Editing; Verma, P.J., Sumer, H., Liu, J., Eds.; Springer: New York, NY, USA, 2022; pp. 99–114. [Google Scholar]
  154. Castaño, J.; Bueno, C.; Jiménez-Delgado, S.; Roca-Ho, H.; Fraga, M.F.; Fernandez, A.F.; Nakanishi, M.; Torres-Ruiz, R.; Rodríguez-Perales, S.; Menéndez, P. Generation and characterization of a human iPSC cell line expressing inducible Cas9 in the “safe harbor” AAVS1 locus. Stem Cell Res. 2017, 21, 137–140. [Google Scholar] [CrossRef]
  155. Lundin, A.; Porritt, M.J.; Jaiswal, H.; Seeliger, F.; Johansson, C.; Bidar, A.W.; Badertscher, L.; Wimberger, S.; Davies, E.J.; Hardaker, E.; et al. Development of an ObLiGaRe doxycycline inducible Cas9 system for pre-clinical cancer drug discovery. Nat. Commun. 2020, 11, 4903. [Google Scholar] [CrossRef]
  156. Bourbon, E.; Ghesquières, H.; Bachy, E. CAR-T cells, from principle to clinical applications. Bull. Cancer 2021, 108, S4–S17. [Google Scholar] [CrossRef]
  157. Uscanga-Palomeque, A.C.; Chávez-Escamilla, A.K.; Alvizo-Báez, C.A.; Saavedra-Alonso, S.; Terrazas-Armendáriz, L.D.; Tamez-Guerra, R.S.; Rodríguez-Padilla, C.; Alcocer-González, J.M. CAR-T cell therapy: From the shop to cancer therapy. Int. J. Mol. Sci. 2023, 24, 15688. [Google Scholar] [CrossRef]
  158. Rojas-Quintero, J.; Díaz, M.P.; Palmar, J.; Galan-Freyle, N.J.; Morillo, V.; Escalona, D.; González-Torres, H.J.; Torres, W.; Navarro-Quiroz, E.; Rivera-Porras, D.; et al. CAR T cells in solid tumors: Overcoming obstacles. Int. J. Mol. Sci. 2024, 25, 4170. [Google Scholar] [CrossRef]
  159. Abou-El-Enein, M. The Fate(s) of CAR T-Cell Therapy: Navigating the Risks of CAR+ T-Cell Malignancy. Blood Cancer Discov. 2024, 5, 249–257. [Google Scholar] [CrossRef]
  160. Dabiri, H.; Safarzadeh Kozani, P.; Habibi Anbouhi, M.; Mirzaee Godarzee, M.; Haddadi, M.H.; Basiri, M.; Alizadeh, Z.; Ebrahimi, M.; Farahmand, L.; Hosseini, S.; et al. Site-specific transgene integration in chimeric antigen receptor (CAR) T cell therapies. Biomark. Res. 2023, 11, 67. [Google Scholar] [CrossRef]
  161. Harris, J.D.; Chang, Y.; Syahirah, R.; Lian, X.L.; Deng, Q.; Bao, X.; Li, H.; Zhang, W.; Chen, Y.; Xu, S.; et al. Engineered anti-prostate cancer CAR-neutrophils from human pluripotent stem cells. J. Immunol. Regen. Med. 2023, 20, 1. [Google Scholar] [CrossRef]
  162. Zhang, J.; Hu, Y.; Yang, J.; Li, W.; Zhang, M.; Wang, Q.; Zhang, L.; Wei, G.; Tian, Y.; Zhao, K.; et al. Non-viral, specifically targeted CAR-T cells achieve high safety and efficacy in B-NHL. Nature 2022, 609, 369–374. [Google Scholar] [CrossRef]
  163. Chen, W.; Tan, L.; Zhou, Q.; Li, W.; Li, T.; Zhang, C.; Wei, X.; Liu, J.; Huang, Y.; Xu, H.; et al. AAVS1 site-specific integration of the CAR gene into human primary T cells using a linear closed-ended AAV-based DNA vector. J. Gene Med. 2020, 22, e3157. [Google Scholar] [CrossRef]
  164. Zhang, Y.; Liu, X.; Zhang, J.; Zhang, C. Site-specific integration of CAR gene into Jurkat T cells with a linear close-ended AAV-based DNA vector for CAR-T engineering. Biotechnol. Lett. 2016, 38, 1423–1431. [Google Scholar] [CrossRef]
  165. Ma, S.; Wang, X.; Hu, Y.; Lv, J.; Liu, C.; Liao, K.; Zhu, X.; Yang, Q.; Chen, H.; Li, F.; et al. Enhancing site-specific DNA integration by a Cas9 nuclease fused with a DNA donor-binding domain. Nucleic Acids Res. 2020, 48, 10590–10601. [Google Scholar] [CrossRef]
  166. Mo, F.; Duan, S.; Jiang, X.; Yang, X.; Hou, X.; Shi, W.; Cueva Jumbo Juan Carlos; Liu, A.; Yin, S.; Wang, W.; et al. Nanobody-based chimeric antigen receptor T cells designed by CRISPR/Cas9 technology for solid tumor immunotherapy. Signal Transduct. Target. Ther. 2021, 6, 80. [Google Scholar] [CrossRef]
  167. Zhen, X.; Kim, J.; Kang, J.S.; Choi, B.J.; Park, K.H.; Lee, D.S.; Hong, S.H.; Lee, J.H. Homology-independent targeted insertion-mediated derivation of M1-biased macrophages harbouring Megf10 and CD3ζ from human pluripotent stem cells. EBioMedicine 2024, 109, 105390. [Google Scholar] [CrossRef] [PubMed]
  168. Zhang, J.; Webster, S.; Duffin, B.; Bernstein, M.N.; Steill, J.; Swanson, S.; Merling, R.; Gonzales, A.; Neumann, C.; Pitt, L.M.; et al. Generation of anti-GD2 CAR macrophages from human pluripotent stem cells for cancer immunotherapies. Stem Cell Rep. 2023, 18, 585–596. [Google Scholar] [CrossRef]
  169. Hale, M.; Lee, B.; Honaker, Y.; Leung, W.-H.; Grier, A.E.; Jacobs, H.M.; Davila, M.L.; Vera, J.F.; Weber, K.L.; Cooper, L.J.N.; et al. Homology-directed recombination for enhanced engineering of chimeric antigen receptor T cells. Mol. Ther. Methods Clin. Dev. 2017, 4, 192–203. [Google Scholar] [CrossRef]
  170. Cetin, B.; Erendor, F.; Eksi, Y.E.; Sanlioglu, A.D.; Sanlioglu, S. Gene and cell therapy of human genetic diseases: Recent advances and future directions. J. Cell. Mol. Med. 2024, 28, e70056. [Google Scholar] [CrossRef] [PubMed]
  171. Bulcha, J.T.; Wang, Y.; Ma, H.; Tai, P.W.L.; Gao, G. Viral vector platforms within the gene therapy landscape. Signal Transduct. Target. Ther. 2021, 6, 53. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, Z.; Zhang, S.; Wong, H.T.; Li, D.; Feng, B. Targeted gene insertion: The cutting edge of CRISPR drug development with hemophilia as a highlight. BioDrugs 2024, 38, 369–385. [Google Scholar] [CrossRef] [PubMed]
  173. Stephens, C.J.; Lauron, E.J.; Kashentseva, E.; Lu, Z.H.; Yokoyama, W.M.; Curiel, D.T. Long-term correction of hemophilia B using adenoviral delivery of CRISPR/Cas9. J. Control Release 2019, 298, 128–141. [Google Scholar] [CrossRef]
  174. Estève, J.; Blouin, J.M.; Lalanne, M.; Azzi-Martin, L.; Dubus, P.; Bidet, A.; Harambat, J.; Llanas, B.; Moranvillier, I.; Bedel, A.; et al. Targeted gene therapy in human-induced pluripotent stem cells from a patient with primary hyperoxaluria type 1 using CRISPR/Cas9 technology. Biochem. Biophys. Res. Commun. 2019, 517, 677–683. [Google Scholar] [CrossRef]
  175. Torres, R.; Garcia, A.; Jimenez, M.; Rodriguez, S.; Ramirez, J.C. An integration-defective lentivirus-based resource for site-specific targeting of an edited safe-harbour locus in the human genome. Gene Ther. 2014, 21, 343–352. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, J.; Exline, C.M.; DeClercq, J.J.; Llewellyn, G.N.; Hayward, S.B.; Li, P.W.; Porteus, M.H.; Gregory, P.D.; Holmes, M.C.; Urnov, F.D.; et al. Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat. Biotechnol. 2015, 33, 1256–1263. [Google Scholar] [CrossRef] [PubMed]
  177. Gomez-Ospina, N.; Scharenberg, S.G.; Mostrel, N.; Bak, R.O.; Mantri, S.; Quadros, R.M.; Gurumurthy, C.B.; Lee, C.; Bao, G.; Suarez, C.J.; et al. Human genome-edited hematopoietic stem cells phenotypically correct Mucopolysaccharidosis type I. Nat. Commun. 2019, 10, 4045. [Google Scholar] [CrossRef] [PubMed]
  178. De Ravin, S.S.; Reik, A.; Liu, P.-Q.; Li, L.; Wu, X.; Su, L.; Raley, C.; Theobald, N.; Choi, U.; Song, A.H.; et al. Targeted gene addition in human CD34+ hematopoietic cells for correction of X-linked chronic granulomatous disease. Nat. Biotechnol. 2016, 34, 424–429. [Google Scholar] [CrossRef]
  179. Kim, D.-H.; Choi, S.-H.; Sung, J.J.; Kim, S.; Yi, H.; Park, S.; Park, C.W.; Oh, Y.W.; Lee, J.; Kim, D.S.; et al. Long-term correction of hemophilia A via integration of a functionally enhanced FVIII gene into the AAVS1 locus by nickase in patient-derived iPSCs. Exp. Mol. Med. 2025, 57, 184–192. [Google Scholar] [CrossRef]
  180. Lyu, C.; Shen, J.; Wang, R.; Gu, H.; Zhang, J.; Xue, F.; Liu, X.; Zhao, W.; Li, Q.; Yang, Y.; et al. Targeted genome engineering in human induced pluripotent stem cells from patients with hemophilia B using the CRISPR-Cas9 system. Stem Cell Res. Ther. 2018, 9, 92. [Google Scholar] [CrossRef] [PubMed]
  181. Lebozec, K.; Jandrot-Perrus, M.; Avenard, G.; Favre-Bulle, O.; Billiald, P. Quality and cost assessment of a recombinant antibody fragment produced from mammalian, yeast and prokaryotic host cells: A case study prior to pharmaceutical development. New Biotechnol. 2018, 44, 31–40. [Google Scholar] [CrossRef]
  182. Schütz, A.; Bernhard, F.; Berrow, N.; Buyel, J.F.; Ferreira-da-Silva, F.; Haustraete, J.; van den Heuvel, J.; Hoffmann, J.E.; de Marco, A.; Peleg, Y.; et al. A concise guide to choosing suitable gene expression systems for recombinant protein production. STAR Protoc. 2023, 4, 102572. [Google Scholar] [CrossRef]
  183. Niazi, S.K.; Magoola, M. Advances in Escherichia coli-based therapeutic protein expression: Mammalian conversion, continuous manufacturing, and cell-free production. Biologics 2023, 3, 380–401. [Google Scholar] [CrossRef]
  184. Zhang, F.; Frost, A.R.; Blundell, M.P.; Bales, O.; Antoniou, M.N.; Thrasher, A.J. A ubiquitous chromatin opening element (UCOE) confers resistance to DNA methylation–mediated silencing of lentiviral vectors. Mol. Ther. 2010, 18, 1640–1649. [Google Scholar] [CrossRef] [PubMed]
  185. Abaandou, L.; Quan, D.; Shiloach, J. Affecting HEK293 cell growth and production performance by modifying the expression of specific genes. Cells 2021, 10, 1667. [Google Scholar] [CrossRef]
  186. Yang, H.; Wang, J.; Zhao, M.; Zhu, J.; Zhang, M.; Wang, Z.; Gao, Y.; Zhu, W.; Lu, H. Feasible development of stable HEK293 clones by CRISPR/Cas9-mediated site-specific integration for biopharmaceuticals production. Biotechnol. Lett. 2019, 41, 941–950. [Google Scholar] [CrossRef]
  187. Tan, E.; Chin, C.S.H.; Lim, Z.F.S.; Ng, S.K. HEK293 cell line as a platform to produce recombinant proteins and viral vectors. Front. Bioeng. Biotechnol. 2021, 9, 796991. [Google Scholar] [CrossRef]
  188. Sun, H.; Wang, S.; Lu, M.; Tinberg, C.E.; Alba, B.M. Protein production from HEK293 cell line-derived stable pools with high protein quality and quantity to support discovery research. PLoS ONE 2023, 18, e0285971. [Google Scholar] [CrossRef]
  189. Leitão, M.d.C.; Cabral, L.S.; Piva, L.C.; Queiroz, P.F.d.S.; Gomes, T.G.; de Andrade, R.V.; Perez, A.L.A.; de Paiva, K.L.R.; Báo, S.N.; Reis, V.C.B.; et al. SHIP identifies genomic safe harbors in eukaryotic organisms using genomic general feature annotation. Sci. Rep. 2025, 15, 7193. [Google Scholar] [CrossRef]
  190. Yuan, M.; Zhang, J.; Gao, Y.; Yuan, Z.; Zhu, Z.; Wei, Y.; Wu, T.; Han, J.; Zhang, Y. HMEJ-based safe-harbor genome editing enables efficient generation of cattle with increased resistance to tuberculosis. J. Biol. Chem. 2021, 296, 100497. [Google Scholar] [CrossRef] [PubMed]
  191. Miyata, Y.; Tokumoto, S.; Arai, T.; Shaikhutdinov, N.; Deviatiiarov, R.; Fuse, H.; Gogoleva, N.; Garushyants, S.; Cherkasov, A.; Ryabova, A.; et al. Identification of genomic safe harbors in the anhydrobiotic cell line, Pv11. Genes 2022, 13, 406. [Google Scholar] [CrossRef]
  192. Butterfield, K.T.; McGrath, P.S.; Han, C.M.; Kogut, I.; Bilousova, G. Generation of an Induced Pluripotent Stem Cell Line with the Constitutive EGFP Reporter. Methods Mol. Biol. 2020, 2155, 11–21. [Google Scholar] [PubMed]
  193. Stellon, D.; Tran, M.T.N.; Talbot, J.; Chear, S.; Khalid, M.; Pébay, A.; Vickers, J.C.; King, A.E.; Hewitt, A.W.; Cook, A. CRISPR/Cas-Mediated Knock-in of Genetically Encoded Fluorescent Biosensors into the AAVS1 Locus of Human-Induced Pluripotent Stem Cells. Methods Mol. Biol. 2022, 2549, 379–398. [Google Scholar]
  194. Stellon, D.; Talbot, J.; Hewitt, A.W.; King, A.E.; Cook, A.L. Seeing Neurodegeneration in a New Light Using Genetically Encoded Fluorescent Biosensors and iPSCs. Int. J. Mol. Sci. 2023, 24, 1766. [Google Scholar] [CrossRef] [PubMed]
  195. Guichardaz, M.; Bottini, S.; Balmas, E.; Bertero, A. Overcoming the Silencing of Doxycycline-Inducible Promoters in hiPSC-derived Cardiomyocytes. Open Res. Eur. 2024, 4, 266. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The criteria for SHS identification in the human genome are evolving alongside advances in genome biology. SHS selection criteria can be divided into (i) foundational criteria based on linear genome features, and (ii) criteria based on 3D genome organization and epigenetic hallmarks. Additional criteria are listed to ensure safety, neutrality, and stability of gene expression (BOX 1), and the current challenges in SHS selection are listed as well (BOX 2).
Figure 1. The criteria for SHS identification in the human genome are evolving alongside advances in genome biology. SHS selection criteria can be divided into (i) foundational criteria based on linear genome features, and (ii) criteria based on 3D genome organization and epigenetic hallmarks. Additional criteria are listed to ensure safety, neutrality, and stability of gene expression (BOX 1), and the current challenges in SHS selection are listed as well (BOX 2).
Cells 15 00081 g001
Figure 2. Approaches for the identification of SHSs. Two main strategies are used: (i) a classical approach involving lentiviral vectors to obtain a single integration event per cell, followed by functional validation, integration site mapping, and safety assessment of candidate loci; and (ii) an in silico approach that employs computational tools to scan the genome for candidate sites fulfilling predefined criteria, followed by site-specific nuclease targeting and functional validation.
Figure 2. Approaches for the identification of SHSs. Two main strategies are used: (i) a classical approach involving lentiviral vectors to obtain a single integration event per cell, followed by functional validation, integration site mapping, and safety assessment of candidate loci; and (ii) an in silico approach that employs computational tools to scan the genome for candidate sites fulfilling predefined criteria, followed by site-specific nuclease targeting and functional validation.
Cells 15 00081 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ahmed, A.; Di Molfetta, D.; Iaconisi, G.N.; Caponio, A.; Singh, A.; Bibi, A.; Dolce, V.; Palmieri, L.; Coppola, V.; Fiermonte, G. Human Genome Safe Harbor Sites: A Comprehensive Review of Criteria, Discovery, Features, and Applications. Cells 2026, 15, 81. https://doi.org/10.3390/cells15010081

AMA Style

Ahmed A, Di Molfetta D, Iaconisi GN, Caponio A, Singh A, Bibi A, Dolce V, Palmieri L, Coppola V, Fiermonte G. Human Genome Safe Harbor Sites: A Comprehensive Review of Criteria, Discovery, Features, and Applications. Cells. 2026; 15(1):81. https://doi.org/10.3390/cells15010081

Chicago/Turabian Style

Ahmed, Amer, Daria Di Molfetta, Giorgia Natalia Iaconisi, Antonello Caponio, Ansu Singh, Aasia Bibi, Vincenza Dolce, Luigi Palmieri, Vincenzo Coppola, and Giuseppe Fiermonte. 2026. "Human Genome Safe Harbor Sites: A Comprehensive Review of Criteria, Discovery, Features, and Applications" Cells 15, no. 1: 81. https://doi.org/10.3390/cells15010081

APA Style

Ahmed, A., Di Molfetta, D., Iaconisi, G. N., Caponio, A., Singh, A., Bibi, A., Dolce, V., Palmieri, L., Coppola, V., & Fiermonte, G. (2026). Human Genome Safe Harbor Sites: A Comprehensive Review of Criteria, Discovery, Features, and Applications. Cells, 15(1), 81. https://doi.org/10.3390/cells15010081

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop