Abstract
For the past decades, gene editing demonstrated the potential of attenuating each of the root causes of genetic, infectious, immune, cancerous, and degenerative disorders. More recently, Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-associated protein 9 (CRISPR-Cas9) editing proved effective for editing genomic, cancerous, or microbial DNA to limit disease onset or spread. However, the strategies to deliver CRISPR-Cas9 cargos and elicit protective immune responses requires safe delivery to disease targeted cells and tissues. While viral vector-based systems and viral particles demonstrate high efficiency and stable transgene expression, each are limited in their packaging capacities and secondary untoward immune responses. In contrast, the nonviral vector lipid nanoparticles were successfully used for as vaccine and therapeutic deliverables. Herein, we highlight each available gene delivery systems for the treatment and prevention of a broad range of infectious, inflammatory, genetic, and degenerative diseases.
Keywords: Lipid nanoparticles, CRISPR-Cas9, viral vector delivery, gene editing
Graphical Abstract

Lipid nanoparticles are assembled by microfluidic mixing of lipids and cholesterol then deployed for nucleic acid cargo cell and tissue delivery.
Introduction
Genome editing can revolutionize how biological processes are harnessed for therapeutic gain. Engineered nucleases facilitate gene-editing serving to improve the diagnosis, prevention, and elimination of infectious, inflammatory, degenerative, cancerous, and genetic diseases. In the last decades, significant progress was made in creating mega nucleases, transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs) 1.. Notably, Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-associated protein 9 (CRISPR-Cas9) used as a genome editing tool has revolutionized genetic manipulations to improve the diagnosis, treatment, and prevention of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), methicillin-resistant staphylococcus aureus (MRSA), human immunodeficiency virus (HIV) excision, stem cell and cancer therapeutics. The promise began in 2007 when Mario R. Capecchi was awarded the Nobel prize in physiology or medicine for this groundbreaking discovery of DNA recombination 2. The specificity of gene editing was improved by introducing double-stranded breaks (DSBs) at the targeted site in dsDNA 3 and by using the host cells’ own DNA-repairing mechanisms 4. Two major DNA repair pathways in eukaryotic cells are, non-homologous end joining (NHEJ, which is error-prone) and homology-directed repair (HDR, which affords accurate gene editing). While the use of CRISPR-Cas9 as a gene-editing tool has come a long way the major limitation in translating it use to the clinic centers on targeted and off target delivery. Approaches available include the use of viral vectors, virus-like particles (VLPs), nonviral polymeric nanoparticles, and lipid nanoparticles (LNPs). Amongst these LNPs elicit excellent cellular uptake (Figure 1). LNPs are also safer than viral vectors with limited immunogenicity. Additionally, the lipid composition of LNPs can be optimized to delivery efficiency as its external surface can be modified for on-target delivery. Such modifications limit off-target toxicities 5.
Figure 1.

Schematic for the LNP delivery of RNA-based CRISPR-Cas9 (sgRNA and Cas9 encoding mRNA). LNPs enter the cells by receptor-mediated endocytosis or hydrophobic interactions. The acidic pH in the endosome anionic interaction with the LNP ionizable lipids and RNAs (Cas9 mRNA and guide RNA) followed by maturation in endosomal environment. These are released into the cytosol following endosomal escape. The Cas9 mRNA is translated to proteins, which forms complexes with the guide RNA (gRNA). The CRISPR-Cas9 complexes are trans-ported to the nucleus. CRISPR-Cas9 activities occur inside the nucleus resulting in the desired gene-editing on targeted locus. In attempts to address therapeutic safety, the review provides a comprehensive review of CRISPR-Cas9-based gene editing on how this can optimize the broad applicability of LNPs including targeted cells and tissue delivery (The figure was created internally by the authors).
Genome Editing Tools
ZFNs, TALENs and CRISPR-Cas9
TALENs and ZFNs are gene editing tools. ZFNs, was the first programmable nucleases, consisting of a non-specific nuclease, FokI (a type II S restriction enzyme from Flavobacterium okeanokoites), fused to a zinc-finger DNA-binding domain 6. The zinc finger proteins are formed by binding tandem arrays of Cys2His2 residues (two invariant pairs of cysteine and histidine amino acid residues) with zinc (II) ions, wherein each zinc finger (ZF) can recognize a 3–4 base pair (bp) sequence 7. The ZFNs bind in the major groove of the DNA double helix and can be modified to identify different DNA sequences 6,8. FokI nucleases cleave the DNA into dimer form; therefore, the ZFNs are designed in pairs to address each target 8,9. The specificity of the ZFNs is primarily determined by the number of fingers in zinc finger proteins and the extent of interaction of the nuclease domain 10. Three- and four-finger ZFNs have been successfully used for the genomic cleavage, targeting 18–36 bp sequences per ZF pair 8. ZFNs have been bioengineered with linkers that allow for increasing the configurations to selectively identify diverse targets 11. ZFNs have been used for various therapeutic applications, including the generation of HIV-resistant genotypes, by disrupting the CCR5 co-receptor in CD4+ T-cells 12. However, the generation of on-target specificity using ZF motifs is costly 7. This has led to the development of TALE-based nucleases. TALENs were developed by fusion of transcription activator-like effectors (TALEs), a protein from Xanthomonas bacteria) with the catalytic domain of the FokI restriction endonuclease 13. Structurally, TALEs consist of an N-terminal, a C-terminal, and a highly conserved 30–35 amino acid residue-long central repeat region. The DNA binding specificity of the TALE proteins is associated with a variable pair of residues located at positions 12 and 13 within each repeat of the central tandem 13. By rearranging or substitution of each residue of each pair, the specificity for a nucleotide can be modified to achieve optimal selectivity. For example, when the residue pair is NG (where N is asparagine followed by glycine), the TALEs specifically bind to thymine. NI (where N is asparagine followed by isoleucine) recognizes adenine, whereas HD (where H is histidine followed by aspartic acid) recognizes cytosine. However, TALENs are best in identifying methylation-dependent gene modifications 14. Analogous to ZFNs, TALENs are also used in pairs. Here FokI cleaves the DNA as a dimer. Apart from FokI restriction endonuclease, the TALEs have been fused with other endonucleases (PvuIII) and meganucleases (I-TevI, I-OnuI). Most do not require dimerization which reduces the number of inter-biomolecular interactions. As these have sequence specificity they limit target diversity 15 and improve specificity 16. TALENs and ZFNs show different mutation patterns. TALEN-induced mutations result in a higher frequency of deletions, whereas ZFNs lead to equal insertions and deletions 17. A notable feature of TALENs is its ability to generate Universal Chimeric Antigen Receptor 19 T-cells (UCART19) through the editing of CD52 and T-cell receptor (TCR). This has been used in targeting B-cell acute lymphoblastic leukemia 18 and demonstrating the translational potential of gene-editing therapy in clinical settings. In this instance, two infants with no feasibility of manufacturing autologous CAR19 T-cells were treated successfully with TALENs for B-cell acute lymphoblastic leukemia 18. Although TALENSs are difficult to deliver this study established proof-of-concept for their use 19. Both are hybrid proteins that have been developed to target and cleave the respective dsDNA. The major disadvantage of ZFNs and TALEN lies in their design, as each new target requires considerable time and labor. In contrast, the CRISPR-Cas9 system is an improvement over the former as it demonstrates design feasibility, reduced cost, and improved specificity 20. Designing a protein is relatively challenging for ZFNs, TALEN-mediated targeted DNA binding compared to the construction of a related CRISPR-Cas9 complex. The latter requires changes only in the single-guide RNA (sgRNA) sequence. As such, the ease in its development makes its usage feasible. Moreover, a recent study which compared the specificity of the three most used gene-editing tools ZFNs, TALENs, and CRISPR-Cas9 in a genome-wide sequencing technique (GUIDE-seq/genome-wide unbiased identification of double-stranded breaks enabled by sequencing) to evaluate any off-target activity revealed that SpCas9 (Cas9 from Streptococcus pyogenes) was more specific, efficient, and safe in gene editing than ZFNs and TALENS. This evaluation was made during in vivo targeting of the human papillomavirus 16 (HPV16) 21. The specificity of SpCas9 is due to its sgRNAs 21,22. CRISPR-Cas9 serves as a “best” gene-editing tool based on simplicity.
CRISPR-Cas gene editing tool
History
The first description of CRISPR was made by iap gene (a 29 bp direct repeat sequence with 32 bp spacers and isozyme of alkaline phosphatase) studies 23. Similar interrupted direct repeats (36 bp direct repeats interspersed by 35–41 bp spacers) are in Mycobacterium tuberculosis 24, archaeal organisms (Haloferx and Haloarcula species), and prokaryotes. All are short regularly-spaced repeats (SRSRs) 25. The term “CRISPR” was introduced in 2002 26,27. Shortly thereafter, the cas genes, present adjacent to the CRISPR locus were discovered. These have known helicase/exonuclease activities with a functional relationship to the CRISPR locus 26,28. A critical insight came when the nucleotide spacers were found to have homology with plasmid and viral genes 29,30 and leading to the notion that CRISPR cascades alter adaptive immunity 31. Integrating the new bacteriophage DNA into the CRISPR array of Streptococcus thermophilus where it recognized a wave of attacking bacteriophage supports the idea that CRISPR affects immunity 32. This supported CRISPR’s role for “genetic vaccination.” The findings spurred studies of spacer sequences derived from invading phages were transcribed into CRISPR RNA (crRNAs), which guided the Cas proteins to its targets 33. It was later discovered that Cas9 can affect cleavage with crRNAs. The discovery of a small RNA, termed trans-activating CRISPR RNA (trcrRNA), which forms a duplex with crRNA and guides Cas9 to its target 34.
A major breakthrough was in the discovery of a programmable single-guide RNA (sgRNA). This combined with trcrRNA:crRNAs highlights the CRISPR-Cas system as a means for RNA-programmable genome editing 4,35. The first successful application of the CRISPR-Cas9 was harnessed by the dairy industry. It was used to immunize cultured bacteria against bacteriophages. Later, CRISPR-Cas9 systems were used in mammalian cells 36,37. Since then, CRISPR-Cas was applied to human, yeast, nematodes, mice, monkeys, rabbits, and other eukaryote cells beyond microbial pathogens 22,36–39. CRISPR has also been used in clinical diagnostic and therapeutic trials (Figure 2).
Figure 2.

CRISPR scissors are applied as nanotheranostics for treatment of emerging infections. Molecular scissors are delivered in vivo by lipid nanoparticles to edit the genome at the desired site. CRISPR nanodiagnostics to diagnose SARS-CoV-2 using lateral flow strips after collecting a nasopharyngeal swab. CRISPR nanotherapeutics can be applied for the inhibition of SARS-CoV-2 and bacterial infections (The figure was created internally by the authors).
Mechanisms of Action
CRISPR systems were classified based on the structural and organizational variations of the Cas genes. Class 1 contains multi-protein effector complexes while class 2 is a single protein. These two classes are further subdivided into six types (type I-VI) with at least 33 subtypes, at which more are continuously being catalogued 40. The class 2 system requires only SpCas9. The SpCas9 protein has two nuclease domains, HNH (named for the characteristic asparagine (N) bookended by single histidine (H) residues) and RuvC-like (named for an endonuclease similar to an E.coli protein) domain, wherein each cleaves the complementary and non-complementary DNA strands 41. For CRISPR-Cas9, the target DNA is followed upstream by a short sequence termed “protospacer adjacent motif” (PAM). The motif is a short 2–6 bp sequence required for Cas9 nuclease activity 4. Approximately ten different CRISPR-Cas proteins were remodeled for gene editing. SpCas9 requires a simple PAM sequence “NGG” (where N is any nucleobase followed by two guanine residues), which presents itself in high frequency at the target DNA. This is to afford the CRISPR-Cas9 system simplicity for its use in diverse applications 41. Mechanistically, the CRISPR-Cas9 system contains two major activity-based components. This includes a DNA-binding domain, that mediates sequence-specific DNA recognition and binding (sgRNA) and an effector domain that enables DNA cleavage. It may also regulate transcription near the binding site (Cas9 endonuclease) 20 nucleotide sequence complementary to the target DNA site and a tracrRNA sequence critical for a unique dsDNA cleavage. The sgRNA associates with the target DNA site by base complementarity, and Cas9 cleaves the dsDNA to generate a double-stranded break (DSB) leads to activation of a native DNA repair mechanism. These repair pathways are exploited for genomic modifications to create either genetic “knock-outs” using NHEJ or genetic “knock-ins” (both for accurate gene editing) by the homology-directed repair (HDR) pathway 41 (Figure 3). Both repair mechanisms occur in many cell types. HDR is triggered only when the homologous sister chromatid is available. In other conditions, where the cells are non-dividing, NHEJ is activated to repair the DSBs 42. The HDR pathway can copy a designed DNA template with the desired sequence inserted into the target cleavage site. NHEJ inhibits gene expression and the gene knock-out process that results in premature stop-codon insertions or frameshift mutations 43. However, when the purpose is to introduce specific mutations and targeted “knock-ins”, DSBs must be repaired using the HDR pathway. The functional efficiency of HDR in comparison to that of NHEJ is relatively low in mammalian cells 44. Methods to improve the HDR efficiency include use of overlapping homology arms, modified Cas9, and intrinsic genetic or chemical modifications, combinatorial approaches were found to improve HDR efficiency. This was done by simultaneously using single-stranded DNA oligonucleotides (ssODN), double sgRNAs, and cell cycle synchronization 46. Importantly, after determining the roles that CRISPR-Cas nucleases play in bacterial defense systems its potential for editing genes in mammalian species were made.
Figure 3.

Mechanism of Cas9 endonuclease to improve efficiency. (A) WT Cas9 endonuclease cleaves the dsDNA after the sgRNA binds to the target DNA, leading to double stranded breaks (DSBs). DNA repair mechanisms introduce random insertions/ deletions (indels) or accurate gene-editing (when homologous DNA is present) that introduce the desired changes, (B) dCas9 (generated by D10A and H48A mutations in WT Cas9), lacks dsDNA cleavage activity and binds specifically with the target DNA that can be used to regulate the transcription of the desired gene. When dCas9 is fused with a transcriptional activator (represented by green sphere), the sgRNA (orange) guides the dCas9 to activate the promoter, thereby resulting in gene expression (CRISPRa). Likewise, attachment of transcriptional repressor (represented by red sphere) results in suppressing the gene expression (CRISPRi), (C) To improve the editing efficiency, deaminase is attached to the nCas9 or dCas9. (CBEs); converts cytosine to uracil followed by DNA repair and replication resulting in conversion of uracil to thymine, thereby encouraging the conversion of C·G to T·A. Similarly, ABEs results in A·T to G·C conversions via deaminating adenine to Hypoxanthine/Inosine. WT=Wild Type, nCas9=Cas9 nickase, dCas9=dead Cas9, CRISPRa=CRISPR activation, CRISPRi=CRISPR interference, ABEs=Adenine Base Editors, CBEs=Cytosine Base Editors, H=Hypoxanthine (The figure was created internally by the authors).
Type of CRISPR Cargos
For CRISPR-Cas9-based genome editing, mainly three forms of cargo choices are available (Figure 4). These cargoes include sgRNA/Cas9 protein complex (commonly referred to as the ribonucleoprotein (RNP) complex); plasmid DNA encoding sgRNA, and Cas9; and RNA system consisting of Cas9 mRNA and sgRNA. The aim of using a cargo is that the functioning Cas9 sgRNA complex must be presented inside cell nucleus for gene editing. The choice of payload often directs which delivery vehicle (described in the delivery section) is to be used for achieving genome editing. This section describes the advantages and disadvantages of each type of cargo and the delivery options available.
Figure 4.

Schematic of plasmid, protein, and mRNA cargoes for CRISPR-Cas9 expression. (A) Plasmid-based expression; two plasmids (encoding Cas9 and sgRNA separately) or a single plasmid (encoding Cas9 and sgRNA together) can be used, (B) Ribonucleoprotein (RNP)-based expression; in vitro transcribed sgRNA can be complexed with Cas9 protein, (C) Cas9 mRNA-based expression; an mRNA encoding Cas9 endonuclease (consisting of 5’ cap, UTRs, and 3’ poly-A tails) along with sgRNA can express CRISPR-Cas9 (The figure was created internally by the authors).
Plasmid DNA
The DNA encoding Cas9 and sgRNA can be packaged in a single or two different plasmids which can be used to express the binary CRISPR-Cas components 47 (Figure 4). Plasmid construction is a time-consuming and labor-intensive process. For the synthesis of Cas9 protein, both transcription and translation are needed, which leads to slow-onset editing; however, plasmids express the Cas9 protein for a longer duration of time. Sustained expression, wherein the goal is to introduce stable mutations can often result in increased off-target activity 48 and a potential risk of insertional mutagenesis 49. Despite its drawbacks identified above, plasmid DNA-based expression is a cost-effective alternative. Another advantage of using plasmid-based expression is the ability to achieve multiplexed gene editing: that is, different sgRNAs can be expressed using a single plasmid, thereby simultaneously ensuring multiple gene site editing and large genomic deletions 50. Moreover, the selective transcription of Cas9 mRNA and sgRNA can be driven by a cell type-specific promoter. Care must be taken in choosing the proper promoter to use; it must be biocompatible with the host cells. For Cas9 expression, the promoter for the β-actin gene in chickens can be used (pX330 and pX458) 38. Recently, studies have focused on using cell-specific promoters that includes macrophage (CD68)-specific promoters. These serve to drive Cas9 expression and can be delivered by nanoparticles 45.
For multiplexing sgRNAs, each sgRNA requires its own promoter for transcription. By doing so, the plasmids can allow for the expression of 2–7 different sgRNA transcripts. 50,51. For the plasmid DNA-based expression of CRISPR components, many delivery options are available. Hydrodynamic injections have been used in mice to deliver the plasmid cargoes. However, this technique is not currently feasible for human use due to the large infusion volume required 52. Also, hydrodynamic injections do not lend themselves to a guarantee that cargo will reach the nucleus of the targeted cells. An electroporation method to deliver cargoes directly to the nucleus of cells during cell division has also been used for plasmid DNA-based delivery in mouse stem cells 48. Different types of lipids like polyethyleneimine (PEI) polyplexes, PAMAM dendrimers, PEI-coated DNA nanoclaws have also been studied to deliver plasmid DNA-based cargo carriers 53, but methods entailing such are not yet suitable for applications in human. The most successful delivery system for plasmid DNA that encodes for CRISPR-Cas9 components has emerged from the use of viral vectors 54. Although plasmid DNA-based delivery for effectuating CRISPR-Cas9 holds tremendous promise, considerable additional research is required to understand any associated off-target activity that results from the stable expression of plasmid DNA encoding Cas9 before clinical applications are considered.
Ribonucleoprotein (RNP)
For the generation of RNPs, Cas9 and sgRNAs (custom-made) can be either purchased or synthesized 55. In research laboratory settings, sgRNAs [generated by in vitro transcription (IVT)] are incubated with the Cas9 protein (which is expressed by E. coli using Cas9-containing plasmids) to generate the RNP complex. The RNP-based delivery of CRISPR components offers several advantages. Since they are ready-for-use protein complexes, no intracellular transcription or translation is required (Figure 4). Accordingly, without consideration to delivery, the efficiency of RNP-based gene editing is superior to that of the plasmid DNA editing 56. The RNP complexes show transient expression due to biological degradation over time, which can help in reducing the likelihood of “off-target” genome editing. However, this same property of RNPs (degradation) renders them as less than ideal for use in cells where stable expression of Cas9 is required. Among the various common delivery methods (physical, viral, and non-viral) available for the trafficking of CRISPR-Cas9, physical delivery, which includes electroporation, are the most efficient for RNP-based expression systems 55,56. Electroporation has been successfully used in cell culture systems and mice zygotes 57. Non-viral carriers for RNP delivery show great promise for high-efficiency gene editing because they allow for the ability to enhance the binding affinity of the Cas9 protein. It was suggested that the Cas9 protein can be modified by attaching negatively-charged tags to improve the interaction of RNP with non-viral-based carriers 55. Recently, the use of a vapor nanobubble (VNB) photoporation technique was used that allows RNP delivery into different cell types. These include primary human T- and mesenchymal stromal cells. They exhibited 60 and 30% of gene knock-out levels in mesenchymal stromal cells and primary human T-cells 58. Although RNP-mediated expression appears promising, more research is required to better understand their immunogenicity and the stability for in vivo delivery.
mRNA
The third type of cargo consists of mRNA encoding the Cas9 nuclease protein. This protein gets directly translated within the cell cytosol. The sgRNA is delivered as synthetic oligonucleotides (Figure 4). As mRNA is susceptible to cell nucleases, various modifications have been made to the chemical constituents comprising mRNA. This enables sustained expression 37. Exogenous mRNA is highly immunogenic and activates multiple pattern recognition receptors (PRRs) and RNA sensors. This includes toll-like receptors (TLRs) 7 and 8, a retinoic acid-inducible gene I (Rig-I), RNA-dependent protein kinase (PKR), melanoma differentiation-associated protein 5 (MDA5), and 2′-,5′-oligoadenylate synthetase/RNase L axis 59,60. The cell immune system recognizes triphosphate, diphosphate, uridine-rich sequences, double-stranded regions, and abnormal 3′- and 5′-cap structures of RNA 59. To prevent the activation of innate immune responses, chemical modification of the exogenous mRNA was considered by adding eukaryotic-mimetic mRNA caps at the terminals of exogenous mRNA and replacing uridine with various bases, including pseudouridine (Ψ), N1-methylpseudouridine (m1Ψ), 5-methylcytidine (m5C), 5-hydroxymethylcytosine (5hmC), 5-methyluridine (m5U), and 2-thiouridine (s2U), 59. Moreover, it was suggested that chemical modifications in the sgRNA oligonucleotides can further enhance efficiency of genome-editing 61. For example, such modifications can be applied to CD34+ hematopoietic stem and progenitor cells and human primary T-cells.
The addition of a 5′ cap with a 3′ poly-A tail to the sgRNA enhances its stability and allows it to exhibit more DSBs in human cells than an unmodified equivalent sgRNA 62. The association of a hairpin secondary structure-engineered sgRNAs (hp-sgRNAs) with five variants of Cas9 exhibited greater specificity in editing the genome at the targeted sites 63. Generation of rationally chemically modified sgRNA improved site-specific genome editing and gene disruption activities in human cells. 2′-O-methylation base change at the 2′-position of the sugar molecule in RNA favors an energetics-favorable conformational change in RNA. Creating sgRNA with modifications such as 2′-O-methylated nucleotides at 5′ and/or 3′ termini of RNA having a phosphorothioate (PS) backbone greatly enhanced CRISPR-Cas9-mediated genome-editing with increased stability and resistance to exonuclease 64. Chemically-modified sgRNA (such as 2′-O-Me- and PS-modifications of three nucleotides at both ends of sgRNA (5′- and 3′-sgRNA) have enhanced gene-editing activity in mammalian cells compared to using unmodified sgRNA  However, in some cases, the addition of 2 to 3 nucleotides at the 5’ end of However, in some cases, the addition of 2 to 3 nucleotides at the 5’ end of sgRNA reduces the formation of the R-loop and cleavage activity of the RuvC domain. Also, the incorporation of a 20 nucleotide RNA hairpin at the 5’ end of sgRNA showed stable R-loop formation with gene-editing activity inactivation 19. Modification of the 5′-end nucleotide of sgRNA to 5-bromo-2′-deoxyuridine (BrdU) in CRISPR-associated Marinitoga piezophila Argonaute (MpAgo) showed higher specificity and affinity to its complementary nucleic acids with a 300-fold enhanced ability to discriminate on-target sequences from off-target sequences that have only one nucleotide difference 66. An in vitro study indicated that exposure of human blood cells to sgRNA with phosphate removal at the 5’ end showed suppression of an interferon (IFN) response 67. In zebrafish embryos, it has been reported that treatment of sgRNAs with a G-quadruplex motif at the 3′ end significantly improved the stability and efficiency of site-specific cleavage of sgRNAs. Such sgRNA with its modified 3’ end did not show any non-specific cleavage in zebrafish 68. In practice, sgRNAs are generated via performing their chemical syntheses, IVT and intracellular transcription in the host cells their advantages, such as affording high purity, relative ease of sgRNAease of sgRNA modification, and high yield 69, 70. However, the limited length of RNAs remains a barrier to their chemical syntheses. Chemical ligation of two or more RNA fragments using click chemistry could be a potential approach towards overcoming the relevant limitations entailed with their chemical syntheses 71.
Various modified sgRNAs that included 2′-O-methyl, 3′-phosphorothioate, 2′-O-methyl-3′phosphorothioate (MS), 2′-O-methyl 3′ thiophosphonoacetate, 4′-O-methyl, 2′-F, locked and unlocked nucleic acids (LNAs and UNAs, respectively), in 3′ and 5′ terminals of sgRNA, increased the editing efficiencies, stability, and on-target sgRNAs performances in human cells. 72–74. In contrast to the limitations in synthetically preparing sgRNAs, the IVT approach that leads to their development is rapid, inexpensive, easy to control, and capable of generating sgRNAs in high yield 69,75. However, sgRNAs might demonstrate poor stability, high immunogenicity, and variable nucleotide sequence lengths. These could result in undesirable secondary or tertiary structures, and thus yield broad heterogeneous activities 62. Several chemical modifications of RNA nucleotides that decrease the immunogenicity and increase the stability have been studied 27. Among various Cas9 nuclease delivery methods, mRNA-based expression is optimal. First, Cas9 mRNA does not need to bypass the nuclear envelope, which in turn, allows for the use of a reduced dosage for achieving efficient transfection. Second, the delivery method does not carry with itself the risk of being integrated into the genome. Third, it can be produced in a cost-effective manner and chemically modified on a larger scale. Most importantly, it is associated with producing limited off-target effects due to its transient expression 76. Owing to the success of COVID-19 mRNA vaccines as well as the existing approaches to gene therapies, current efforts disproportionately favor the use of Cas9 mRNA along with sgRNA oligonucleotides as a cargo for the delivery of CRISPR components against multiple pathogens 45,77,78. Despite the advances in CRISPR-Cas9-based gene editing, concerns exist, some of which are discussed in the next section.
Limitations of CRISPR-Cas9
Progress for CRISPR-Cas9 was realized in genetic, cancerous, and infectious disease treatments 79. Though the approach has seen favorable results in pre-clinical research studies the promise of gene editing in clinical trials has yet to be achieved. With regards to clinical trial usage, concerns related to the specificity, efficiency, and in vivo delivery of CRISPR-based gene-editing therapies must first be resolved for their safe and efficient clinical translation.
Specificity
Nucleotide targeting specificity has been a significant concern for each of the ZFNs, TALENs, and CRISPR-Cas9-based systems. CRISPR-Cas9 enzymes leading to the dsDNA breaks at off-target locations (in addition to the targeted sites) can result in undesirable off-target activity 37. The Cas9 enzyme sometimes cleaves dsDNA that does not exactly complement the designed sgRNA, thereby leading to mutations at non-targeted sites. Long-term expression of Cas9 within the targeted cells, editing temperature, and unsuccessful sgRNA design are some of the factors that can significantly impact off-target activity 21. Off-target prediction assays including in silico, biochemical, and cell-based systems were developed to model these unwanted mutations 80. Some of the examples of off-target prediction assays include GUIDE-seq (genome-wide unbiased identification of DSBs enabled by sequencing) 81, LAM-HTGTS (linear amplification-mediated high-throughput genome-wide translocation sequencing) 11 and CIRCLE-seq (circularization for in vitro reporting of cleavage effects by sequencing) 82. In parallel, studies are being conducted to address the challenge of producing off-target effects caused by wild type SpCas9.
Highly specific Cas9 variants of wild-type SpCas9 have already been identified 21. A variant protein called cas9 nickase (nSpCas9) was generated by mutating either the HNH (D10A)- or RuvC (H48A)-like domain, resulting in conferring it the ability to cleave single-stranded DNA (ssDNA), wherein it has been demonstrated that nicks in the genomes are repaired with high fidelity as compared to DSBs 83. Likewise, introducing mutations in both HNH (D10A)- and RuvC (H48A)-like domains results in the generation of a mutant protein called dead cas9 (dCas9), which binds with the sgRNA and targets DNA with high specificity. dCas9 lacks DNA cleavage activity, and thus can essentially deliver any functional domain to a specific locus in the genome. Such delivery can allow for site-specific transcriptional activation (CRISPRa) or transcriptional inhibition (CRISPRi) of the desired gene 84. To improve the efficacy of CRISPRi, dCas9 was fused with a repressive chromatin modifier domain, kruppel-associated box (KRAB) from kox1 protein, recruiting chromatin-modifying complexes to silence the transcription of genes targeted by sgRNA 85. Moreover, SpCas9 protein can be easily programmed for multiplexing, which means multiple genomic loci can be targeted by several sgRNAs simultaneously to further increase specificity 65.
Efficiency
dsDNA cleavage by CRISPR-Cas9 results in the activation of repair pathways in the edited cells, which in turn, leads to gene editing. The two most common pathways are NHEJ and HDR, wherein the latter results in gene editing with higher accuracy. However, the editing efficiency of the HDR repair pathway is low 62. The strategies to improve the HDR repair efficiency by inhibiting the NHEJ pathway was described. This includes regulating HDR-related factors and by synchronizing the cell cycle 21. To improve the editing efficiency base-editing tools were developed that allow the irreversible conversion of one targeted base to another without introducing dsDNA cleavage 79. Two classes of Base Editors (BEs) have been developed. This includes adenine base editors (ABEs) that convert A·T to G·C 86 and cytosine base editors (CBEs). Each result in the transition of C·G to T·A 87. BEs are comprised of the Cas9 nickase or dCas9 fused to a deaminase and a guide RNA to direct the Cas9 protein to the target base within the specified window. The use of base editors has resulted in 70 and 100% editing efficiencies in mouse neurons and cultured mammalian cells, respectively 21. Base-editing techniques have been widely adapted in prokaryotes, mice, monkeys, and human embryos 21. Further advances were made by establishing the prime editing (PE) technique. PE mediates all the possible base-to-base conversions as well as targeted deletions and insertions with increased accuracy 88. The prime editing complex consists of prime editing guide RNA (pegRNA) that identifies the target sequence by sgRNA and contains the information to replace the targeted sequence in the reverse transcriptase template sequence, wherein it also has a primer binding site (PBS) that is required to start reverse transcription at the 3’ end of the nicked DNA; and a Cas9 nickase fused to reverse transcriptase 89. PEs do not induce detectable off-target effects efficient delivery was optimized.
Overview for Gene Delivery
Delivery of gene-editing tools is a daunting task. Recently, technologies were used in delivering CRISPR-Cas9 components uses non-viral and viral systems. The methods include transfection by lipids, cationic polymers, or lipid nanoparticles (LNPs), electroporation (nucleofection), and transduction. For cell delivery, physical methods such as lipofectamine-mediated transfections and electroporation of plasmid DNAs encoding SgRNAs and Cas9 proteins have been widely used 19,57. However, for in vivo applications, electroporation-based techniques for their delivery are not suitable. Instead, viral-based vectors, including the adeno-associated virus (AAV) 14 and lentivirus, have been used in the past few years. These viral vectors have limited future opportunities for vast applications, due to their immunogenicity, limited payload size, replication, chances of active mutation and carcinogenesis, which are significant safety concerns and fully rollout for genetic medicine. Cas9 orthologues like Staphylococcus auricularis Cas9 (SauriCas9; 1061aa), Staphylococcus aureus Cas9 (SaCas9; 1053 aa) instead of SpCas9 (1350 aa) have been used 79 to overcome the drawback of the low packaging capacity of AAV. Nevertheless, the in vivo delivery of CRISPR-Cas systems is concerning. In consideration of the barriers to effective and efficient delivery, we devote considerable attention and offer insight into the advantages of delivery methods for successful CRISPR-Cas9-based gene-editing.
Delivery Systems
Viral Vectors
Significant technological advancements have been made in the field of gene-editing with the use of viral-based vectors as delivery vehicles. An early mention of using viruses as vectors was in the early 1980s when the inherent capacity of viruses for transducing tissues and cells with foreign nucleic acid was made for gene delivery 91. Throughout the years, viral vectors were developed based on their projected efficacy for delivery and expression 92. Lentiviruses, adenoviruses, and adeno-associated viruses (AAVs) (Figure 5) are common vector systems. They are comprised of a protein capsid used for cargo encapsulation, the transgene of interest, and the promoter regions 93. Their delivery abilities are based on systemic administration. AAV vectors are used for in vivo applications; whereas lentivirus vectors are used for ex vivo purposes 94. Viral vector systems are widely employed due to their high efficacy (John Wiley and Sons LTD, 2021) (Figure 5). However, deleterious effects can result due to immune responses. Pre-existing immunity, limited cargo capacity and potential for insertional mutagenesis reduce efficacy. Alternative non-viral delivery may improve safety, efficiency, and scalability 95. Moreover, viral-based vectors require high viral particle quantities for efficient gene editing 96.
Figure 5.

Viral vectors used for gene delivery. Comparison of the three most-widely used viral vectors: adenovirus (AVs), adeno-associated virus (AAVs) and lentivirus. Total number of currently active clinical trials that employ viral vectors and its schematic representation (figure concept adopted from John Wiley and Sons LTD. (2021) (The figure was created internally by the authors).
Lentiviruses
Lentiviruses are a genus in the Retroviridae family that are approximately 100 nm in diameter. They have enveloped spheres containing two sense-strand RNAs that are encapsulated by nucleocapsid proteins. Reverse transcriptase is present in these viruses along with integrase and protease proteins. Lentiviruses fall into the category of complex retroviruses. HIV-1, a prominent lentivirus, possesses a 9.7 kb genome flanked by 5’- and 3’-Long Terminal Repeat (LTR) regions that are crucial for the replication cycle. The three essential core protein genes have the following functions: (i) the gag gene is ultimately responsible for originating the matrix proteins and the capsid; (ii) the pol gene encodes for reverse transcriptase and integrase; and (iii) the env gene encodes for glycoproteins associated with cellular tropism of the virus 97. Additionally, the tat and rev genes, which are present in lentivirus, are associated with transcription, elongation, and nuclear export of viral RNA 98,99. Moreover, auxiliary genes such as vif, vpu, vpr and nef, in lentiviruses increase pathogenesis and viral titer 100. The most prominent advantage of using lentiviruses in gene-editing applications is their ability to integrate into the host genome and exhibit extended transgene expression. Lentivirus can form replication-competent retroviral vectors (RCR), which pose a risk of large deletion or insertional mutagenesis as dysregulation in viral replication will occur due to RCR. The FDA mandates thorough testing for the detection of RCRs in lentivirus products, a process that is both very expensive and time-consuming. PCRs need to be performed on patients’ specimens from time to time for those participating in clinical trials, which increase burdens on both the patients and on the trial 101. Chimeric Antigen Receptor-T (CAR-T) cells developed from lentivirus for acute lymphoblastic leukemia give rise to B-cell aplasia due to sustained CAR T-cell effect. It can result in B-cell aplasia for more than 5 years following treatment due to the long half-life of T-cells. 102 Late onset of malignancies is also a risk factor for patients receiving lentivirus vector treatments, as proto-oncogenes might be activated near the integration site 103. Moreover, the sites of integration are difficult to predict, which adds to the challenges of utilizing lentiviruses as a therapeutic delivery vehicle (Figure 5). Although multiplexable methods, such as CRISPR - enhanced viral integration site sequencing (CReVIS-seq), for mapping the viral-integration sites have been developed, they are limited to needing specific sections of virus-host boundary sequences to work effectively 104. A single lentivirus vector was demonstrated to express a number of genes 105, which may not be safe for patients long term. As mentioned earlier, the provirus may also integrate into the host genome, thereby causing a cascade of pathogenic reactions. Moreover, there may also be mobilization of structural viral genes in the target cells. Lentiviral vectors while being developed post additional questions on whether they will be safe enough as an effective in vivo therapy 101. They are modified from HIV-1 and while several viral genes are removed concerns remain for recombination. The lentiviral vector contains the viral LTRs and the packaging signal. While the lentiviral packaging genes are contained in separate plasmids and the emerging pseudo lentiviral particles remain replication deficient whether their longer-term use could have secondary untoward outcomes remains undefined. On balance and to date lentiviral vectors are used to deliver genes into cells as to date they are both a dependable and safe method as a gene delivery system.
Adenoviruses
Adenoviruses (Family: Adenoviridae) are DNA viruses without an envelope. They contain an icosahedral protein capsid that can accommodate a 26- to 45-kb double-stranded, linear DNA genome. About 51 different serotypes of adenoviruses are present in humans, from which Ad5 and Ad2 are most naturally occurring. Nearly 12 different proteins make up the virion, including core proteins, terminal proteins, major and minor capsid proteins, maturation protease and packaging machinery. The icosahedral structure is formed by the major coat protein hexon and the capsomers are formed by penton base and fibers. Three minor coat proteins conserved within the family are proteins I, IIIa, V, and VIII, with each playing a pivotal role in the viral life cycle. The Ad genome is packaged within a icosahedral capsid having a diameter of 95 nm measuring from vertex to vertex 106. Ad genome is flanked by two short, inverted terminal repeats that act as the origin of replication 107. Inverted Terminal Repeat (ITR) forms a hairpin-like structure that serves as a self-primer to enhance primase-independent DNA synthesis along with easing its integration into the non-host genome. Proper viral packaging is carried out by a genetic component following the ITR in the Ad genome 65. The Ad contains core proteins V, VII, and µ, which are highly basic and play a role in neutralizing the negative charges to allow for the dsDNA to condense within the viral core 108. AVs while efficient for gene delivery, they do not uniformly target disease affected tissues. Their toxic effects do limit the vector amount that can be administered to patients. Limitations in viral titers and packaging efficiencies also limit their general utility for treating more chronic disease. Nonetheless and on balance, AVs are effective as vectors because of their epichromosomal persistence in host cells, high efficiency of transduction in dormant and dividing cells, and its broad tropism 65.
There are three generations of viral vectors: The first-generation vectors are devoid of the E1 and E3 regions and can package a payload maximum of approximately 8.2 kb of cargo carrier gene(s). The second-generation vectors, along with lacking E1 and E3 regions, also lack the E2 and E4 regions. The third-generation vectors are known as ‘gutless’ vectors that contain only the ITRs and the packaging signal. Also known as ‘high capacity’ adenoviral vectors (HCAds) 109, these third-generation vectors have packaging capacity of approximately 36 kb of genetic material. Therapeutic Ad vectors have been used in vivo since the early 1990s with the first human trials in patients afflicted with peripheral vascular disease and lung tissues of patients with cystic fibrosis 38,110. However, numerous pathologies have been reported in pre-clinical and clinical trials. In 2002, a patient fatally suffered from multisystem organ failure upon involvement in a clinical trial for Ornithine transcarbamylase deficiency (OTCD), with other participants experiencing an acute rise in hepatic transaminase, hypophosphatemia, and thrombocytopenia 111. Delivery of SpCas9 using Ad vector in targeting gene Pten (which is associated with nonalcoholic steatohepatitis (NASH)), was shown to cause liver damage (hepatomegaly and steatohepatitis) and cellular and humoral immunity. 112. Li et al. reported 13% editing was achieved in vivo in red blood cells (RBCs) of the edited hematopoietic/stem progenitor cells that were performed via using the CRISPR-Cas9 in B-thalassemia mice 113. Intravenous administration of Ad5 for Cas9 system targeting mutated human SERPINA gene [encodes for alpha-1-antitrypsin (AAT)], showed the possibility of the use of gene editing in hepatocytes. 114 Efficient tumor regression was reported by “knocking out” mutated oncogenes such as KRAS or EGFR by CRISPR-Cas system 115,116. The heterogeneity of different cancers, along with their tendency to metastasize or diffusely spread, may result in the therapy being less effective than if such was otherwise. A challenge in Ad vector delivery is that the common human Ad serotypes have pre-existing immunity in the form of neutralizing antibodies (NAbs) against them. Seroprevalence of 35% and 90% against HAd5 was found in the United States and Cote d’Ivoire (West Africa), respectively 117. Also, adenoviruses have high clearance rates post-systemic administration due to their interaction with myeloid cells.
Adeno-Associated Viruses (AAVs)
AAVs was a serendipitous discovery in 1965, wherein such was first deemed as an impurity in adenovirus preparations 118. AAV is classified as a dependoparvovirus because it requires a ‘helper virus’ in assisting the completion of its life cycle. AAVs are non-enveloped and are smaller than Ad. They contain a single-stranded genome of 4.7 kb that is flanked by 2 ITRs. These ITRs are 145 nucleotides long and form a hairpin structure that can self-prime 119. The three major genes encoding for a functioning AAV are the Cap gene that encodes for the Capsid consisting of 60 outer-coat proteins responsible for protecting the genomic DNA and cell binding; the Aap gene which encodes for an assembly-activating protein that acts as a scaffold for capsid assembly; and the Rep genes that are responsible for viral packaging and replication. The Rep genes Rep 52, Rep 40, Rep 68, and Rep 78 are present in the first Open Reading Frame (ORF). However, they lack the required genes needed to replicate their genome 120. The capsid of AAV is a T=1 icosahedral, 60-mer structure containing VP1, VP2 and VP3 in a 1:1:10 ratio, respectively. The capsid sequence dictates the varying transduction efficiencies in different tissues 121. Since the advent of AAVs for usage in humans [which afforded the in the delivery of the cystic fibrosis transmembrane regulator (CFTR) gene packaged with the recombinant AAV2 capsid (rAAV2-CFTR)], AAVs have been widely used to deliver a gene of interest in vivo 122. Because of the recent advances in discerning host-virus interactions, recombinant AAV (rAAV) is becoming the leading platform for delivering genes of interest for treating different human diseases 62. AAVs are comprised of an icosahedral protein capsid of ~26 nm in diameter having a single-stranded DNA genome of ~4.7 kb in length that can be either the plus (sense) or minus (anti-sense) strand 123. The small size of the AAV genome limits insert size 93. This aspect especially poses a unique challenge to deliver the CRISPR-based cargos. A dual-vector system using ITR-mediated recombination was exploited to overcome this challenge 124. However, the genome-editing efficacy of this dual-vector approach is potentially limited in its extent of overall success, as accurate editing can be achieved if and only if both vectors are taken up by and expressed in the given cells in a manner that affords them a period of their contemporaneous presence. One other important positive aspect of using an AAV-based CRISPR-Cas9 delivery system is to limit unwanted outcomes. This is affirmed because of its long-term expression of its genome editing abilities. Many approaches towards achieving transient expression of the genome editing system have been reported, including a drug-inducible promoter for gene expression, protein activation by light, drug-inducible assembly, reconstitution from a split construct, and protein inactivation by a drug-inducible destabilizing domain 22,125,126. To date, their clinical efficacy has yet to be demonstrated. Immunogenicity is a major issue to contend with in using AAV viral vector-based delivery systems, especially if requiring the use of a high dose 127 (Figure 5). AAV packaging capacity is lower than that of other viral vectors, thereby at times preventing full-length BEs from being packaged into the vector 128. Maurya et al. wrote that AAV-mediated therapy is “relatively safe” at a low threshold of vector dose 129 with efficacy being mostly dependent on the dose administered 130. This could imply the potential endpoint limits for using AAVs in vivo at “safe” doses. With that stated, it is necessary to emphasize that the efficacy of AAVs cannot be interpreted from in vitro studies because the transduction of AAVs in vivo does not positively correlate with that obtained when applied in vitro. In short, even though AAVs have undergone extensive testing in pre-clinical studies, their translation for human use is still lacking 120.
Virus-like Particles (VLPs)
Virus-like particles are a class of delivery vehicles that resemble the structure of true virus particles, wherein the immune system rapidly recognizes them 131. VLPs address certain drawbacks of viral-based delivery systems: (i) the low likelihood of integration of viral vectors into the genome of transduced cells; and (ii) an increase in off-target editing frequency due to extended expression in transduced cells 132. VLPs can have enveloped or non-enveloped structure with one to four proteins in its outer layer. VLPs are self-assembling structures that contain the wild-type genome. In a study, the therapeutic cargo carrier such as base editing machinery, was linked with the gag polyprotein of Friend Murine Leukemia Virus (FMLV) to make FMLV based VLPs 132.
VLPs and their Types for Gene Therapy
There have been numerous attempts to develop effective VLP-based gene editing systems. Choi et al. demonstrated that pre-packaging the Cas9 protein in lentiviral particles reduced off-target editing because the exposure and expression was transient 133. The first report of in vivo gene editing using VLPs was by engineered VLPs, “eVLPs”. Nanoblades, which are Cas9-sgRNA ribonucleoproteins produce efficient genome editing in in vitro systems such as bone marrow-derived macrophages and primary human fibroblasts. Fusion of Cas9 from Streptococcus pyogenes to Gag from MLV allowed the incorporation of the Cas9 endonuclease into VLPs internal structure. Nanoblades were more effective in inducing DSBs in vitro compared to lipofection or electroporation. They can also be made into complexes with templates of DNA repair to facilitate homologous recombination-based “knock in” in cells without transfection. The subsequent design of eVLPs with the fourth generation eVLPs afforded a 16-fold greater packaging capacity than their first-generation equivalents. Off-target editing was very low, and they found that it was efficacious in a myriad of cell types. This also yielded effective base editing in brain, liver and other organs of mice. Base editing mediated by Pcsk9 had greater than 60% efficiency following a single intravenous dose. eVLPs have been reported to be of some utility in a myriad of both ex vivo and in vivo applications. It is worth noting that the modular structure of eVLPs provides countless number of opportunities for delivering proteins or therapeutic agents of interest, but a priori knowledge of the applicable cargo carrier size that can efficiently diffuse out from these capsids does beget additional consideration 134.
Nonviral Vector
Nonviral delivery vehicles are emerging as promising alternatives for gene delivery due to their improved safety profile, scalability, and versatility. These vectors offer advantages such as lower immunogenicity and reduced risk of insertional mutagenesis compared to viral vectors 135. Non-viral vectors can be categorized into different types. These include, but are not limited to, lipid- and polymer-based vectors. Lipid-based vectors that include liposomes or lipid nanoparticles (LNPs) can encapsulate and protect the therapeutic genetic material. They offer efficient cellular uptake and endosomal escape that enable successful gene delivery. Lipid-based vectors can also be optimized to enhance stability, transfection efficiency, and targeting (Figure 6). Alternatively, polymer-based vectors utilize synthetic polymers, such as polyethyleneimine (PEI) or poly(lactic-co-glycolic acid) (PLGA), to form complexes with DNA or RNA. The complexes protect the genetic material from degradation and facilitate cellular internalization. Lipid and polymer-based vectors can be modified to controlled release and create specific delivery by surface modifications. Non-viral vectors hold potential for clinical translational research. Ongoing advancements in vector design, formulation optimization, and delivery serve to enhance efficiency, safety, and therapeutic potentials 136 137.
Figure 6.

Graphic description of currently developed nanocarriers for in vivo gene editing. A description of the components of this description are now provided. A micelle is defined as a self-assembled, colloidal structure formed by the aggregation of amphiphilic molecules in a liquid medium. Amphiphilic molecules possess both hydrophilic and hydrophobic regions within the same molecule. In an aqueous environment, these molecules arrange themselves spontaneously to minimize exposure of their hydrophobic regions to water. The hydrophobic tails of the amphiphilic molecules come together in the center, shielded from water, while the hydrophilic heads face outward, interacting with the surrounding aqueous medium. Micelles have a core-shell structure, with the hydrophobic core and the hydrophilic shell. This unique arrangement allows them to solubilize and transport hydrophobic materials in a biological aqueous environment. They are a spherical vesicle composed of lipid bilayers analogous to cell membranes with an aqueous core. Liposomes are artificially prepared consisting of phospholipids, the primary building blocks of cell membranes. The phospholipids have hydrophilic heads and hydrophobic tails which enable them to form a bilayer structure in an aqueous environment. Cationic lipids have a positive charge due to the presence of quaternary ammonium groups in their structure. When mixed with negatively charged nucleic acids, electrostatic interactions occur, leading to the formation of lipoplexes. Lipoplexes are widely used in gene delivery and gene therapy applications. (The figure was created internally by the authors).
Targeted Gene Delivery
Nanoparticles (NPs) can be made to better protect genetic cargo carriers of in vivo elimination which occurs via normal excretion processes and metabolic processes. Based on the objective of the therapy, NPs can be modified enabling all kinds of routes of administration. Additionally, the efficacy of large-size cargo carrier materials of the NPs encases and affords its delivery to targeted cells and tissues. The chemical basis and any chemical modifications thereof of NPs can alter their biodistribution, ability to afford cellular and tissue targeting, and the mechanism affording cellular absorption such that the desired therapeutic result is achieved 138.
Polymeric nanoparticles (PNPs)
New drug delivery systems are becoming increasingly important due to the intricacy of some diseases and the toxicity associated with some medicines 139. PNPs can increase the bioavailability and biodistribution of therapeutic payloads 140 and have proven effective for targeting brain subregions 141 and improve drug solubility, release, distribution, limit immunogenicity, and can deliver multiple drugs simultaneously 141,142. Toxicities that are governed by their chemistry, size, shape, aggregation, and electromagnetic characteristics 143–145 can be overcome by design and exposure limitations 146. Delivery of nucleic acids to cells in the form of plasmid DNA (pDNA) or small interfering RNA (siRNA) can improve outcomes 147. Viral-based vectors allow high rates of gene transfer but may induce inflammatory and immunological responses. To mitigate these issues, non-viral-based cargo carriers that included cationic liposomes or polymers were developed. On doing so, they form “lipoplexes” or “polyplexes” (Figure 6). These complexes show promise as an alternative strategy to viral-based vector gene therapy 148. First, a micelle is a self-assembled, colloidal structure formed by the aggregation of amphiphilic molecules in a liquid medium. Amphiphilic molecules possess both hydrophilic and hydrophobic regions within the same molecule. In an aqueous environment, these molecules arrange themselves spontaneously to minimize exposure of their hydrophobic regions to water. The hydrophobic tails of the amphiphilic molecules come together in the center, shielded from water, while the hydrophilic heads face outward, interacting with the surrounding aqueous medium. Second, micelles have a core-shell structure, with the hydrophobic core and the hydrophilic shell. This unique arrangement allows them to solubilize and transport hydrophobic materials in a biological aqueous environment. They are a spherical vesicle composed of lipid bilayers analogous to cell membranes with an aqueous core. Third, liposomes are artificially prepared consisting of phospholipids, the primary building blocks of cell membranes. The phospholipids have hydrophilic heads and hydrophobic tails which enable them to form a bilayer structure in an aqueous environment. A lipoplex is a complex formed by combining lipids (usually cationic lipids) with nucleic acids, such as DNA or RNA. Fourth, cationic lipids have a positive charge due to the presence of quaternary ammonium groups in their structure. When mixed with negatively charged nucleic acids, electrostatic interactions occur, leading to the formation of lipoplexes. Lipoplexes are widely used in gene delivery and gene therapy applications.
Genetic cargo within endosomolytic polymeric carriers is protected by reversible covalent modifications, cellular receptor ligands, and labile siRNA conjugations. Each can be integrated into dynamic poly-conjugate technologies. A significant advancement in the use of RNA interference (RNAi) in therapeutic applications and gene function research would be the affording of efficient in vivo transport of siRNA to the proper target cell. Dynamic poly-conjugate technology has key features that include a membrane-responsive polymer, reversible masking of polymer activity until it reaches the acidic environment of endosomes, for example, targeting hepatocytes in vivo after simple, low-pressure IV injection 149. RNAi, target degradation by RNase H-mediated cleavage, splicing manipulation, non-coding RNA inhibition, gene activation, and programmed gene editing are all examples of using oligonucleotides to influence gene expression. As a result, these molecules could be used to treat a wide range of diseases, especially as numerous oligonucleotide-based medicines have only recently received regulatory approval. Despite recent technological breakthroughs, effective oligonucleotide delivery, particularly to extra-hepatic regions, remains a key translational challenge 150,151. Although NP delivery systems can be used to deliver genes, there are still some challenges that need to be addressed to improve their effectiveness.
mRNAs
Researchers worldwide have been investigating different technologies to develop mRNA delivery systems for more than 30 years. LNPs are novel biopharmaceutical drug delivery systems composed of different classes of lipids. LNPs as a drug delivery vehicle were first approved in 2018 for the siRNA drug. LNPs are found to be the breakthrough nonviral-based technology that can stably deliver mRNA to the target organ, especially due to protecting the mRNA from nuclease-mediated degradation 152. mRNA LNPs have recently come under the spotlight due to serving as remedial platforms following the acceptability of the SARS-CoV-2 mRNA LNP vaccines (BNT162b2 and mRNA-1273), with mRNA vaccine doses now injected into billions of individuals worldwide. These semi-effective mRNA-LNP vaccines have showcased the modularity and the rapid manufacturing capabilities of mRNA-LNP systems 153.
LNPs are lipid-based nanostructures, composed of multiple components such as cholesterol, a phospholipid, a PEG-lipid (polyethylene glycol lipid), and an ionizable lipid. Each type of lipid has a distinct role in typical LNPs formulations and formation. For example, ionizable lipids are responsible for mRNA binding and LNP core formation. Cholesterol and phospholipids are considered helper lipids that give stability to the mRNA-lipid complex. PEGylated lipids protect the lipid shell and the LNPs from opsonization 154 (Figure 7). LNPs can be classified into different subgroups based on lipid composition and the synthetic method used for their development 155. For instance, liposomes, exosomes, transferosomes, nanostructured lipid carriers, liposome-like structures or LNPs, solid LNPs (SLNPs), and lipid nanoemulsions are some of the well-developed lipid-based NPs to date 156. LNPs excel in efficacy over that of viral-based vectors due to their decreased immunogenicity, the use of versatile payloads (from several nucleotides to several million in length), tailorable functional groups, and ease of scale-up which enables commercial production 157. The types of lipids used in mRNA-LNP formulation and methods for synthesis are discussed below.
Figure 7.

mRNA delivery by LNPs. Key components of mRNA-LNPs; administration routes of mRNA-LNP formulations; advantages of mRNA-LNPs over other delivery systems; mRNA-LNPs used in different diseases (The figure was created internally by the authors).
Lipids used in mRNA LNPs Preparation
Cationic Lipid Nanoparticles (cLNPs)
Cationic lipids have three distinct components that include a positively charged (hydrophilic) amine functional group; a hydrophobic chain; and a covalent linkage between the hydrophobic chain and the hydrophilic moiety-linker group. The positively-charged amine group (ammonium based) can readily associate with the negatively-charged nucleic acid through opposing charges interactions, which makes transfection of LNPs effective 158. The hydrophobic moiety includes the length of alkyl chain and its degree of saturation. The polar hydrophilic headgroup positively charged made through the protonation of one or multiple amino groups designed with specific spatial properties. This headgroup contain amines, quaternary ammoniums, guanidiniums, amino acids, peptides, and a variety of heterocyclic compounds. 159. Headgroups with one or multiple positive charges can attract negatively charged nucleic acids. This occurs through electrostatic forces that forms a complex called a “lipoplex” made up of cationic lipids and anionic nucleic acids. This structure is crucial for the cLNPs function as it protects the nucleic acids from degradation and delivers them to target cells. Prior studies have found that the acid dissociation constant (pKa) of the headgroup and the position of the charge on the lipid bilayer interface are key factors for the successful delivery of nucleic acids. 160. Recently, research was performed that confirmed the acid dissociation constant (pKa) of liposomal nanoparticles and showed that they possess the ability to transport siRNA across biological barriers and ultimately achieve gene silencing 161. The presence of a charge on the lipid can cause toxicity and quick elimination from the bloodstream. To address this issue, anionic lipids with pKa values of around seven or lower have been developed, which have lower toxicity and can efficiently encapsulate nucleic acids for both in vitro and in vivo activities as they exist in an uncharged state 162. Cationic lipids are the optimal carrier for gene delivery, as they can be easily synthesized by modifying each of the constituent domains of the ionizable lipids 159. The cationic lipids that are used as part of non-viral-based gene carriers include N-(1-(2,3-dioleyloxy) propyl)-N-,N-,N-trimethyl-ammonium (DOTMA), 1,2-dioleoyloxy-3-(trimethylammonium) propane (DOTAP), 2,3-dioleyloxy-N-(2-(sperminecarboxamido)ethyl)-N,N-dimethyl-1-propana trifluoroacetate (DOSPA), DC-cholesterol (dimethylaminoethane-carbamoyl-cholesterol) and 1,2-dimyristyloxy-propyl-3-dimethyl-hydroxyethylammoniumhydroxyethyl ammonium bromide (DMRIE). Recently, various polymeric carriers, such as chitosan, poly(β-amino esters) (PBAEs), protamine (when used only with polyethyleneimine (PEI), and poly(2-dimethylaminoethyl methacrylate) (PDMAEMA) and other polyamines have been adapted from their use in siRNA/DNA delivery to now afford mRNA delivery 163. A linear PEI derivative, jet PEI, which is commonly used for DNA and siRNA transfection, can also be used for mRNA transfection, even though it shows lower efficiency than lipoplexes 164. LNPs have been reported to reduce the susceptibility of nucleic acids to degradation by nucleases. Modifying the ratio of cationic lipid to nucleic acids, by having more positive charge, improves nucleic acid delivery by making it easier to bind to negatively charged cell surfaces and break down endosomal membranes. Despite their relatively low transfection efficiency when compared to that of using viral-based vectors, LNPs can be used in a wide range of applications due to the rapid and facile preparation of lipoplexes whilst displaying relatively low toxicity 165. However, cationic LNPs systems tend to degrade when stored for a long time in aqueous solutions, particularly due to their tendency to undergo hydrolysis. 166. The physical and chemical properties of cationic lipids greatly hinder their cellular uptake and effectiveness in gene therapy. One study observed that the lipoplex size is the primary factor in determining the efficiency of lipofection in vitro 167. Furthermore, studies have indicated that larger liposomes are rapidly cleared from the bloodstream by the reticuloendothelial system (RES), while smaller liposomes exhibit prolonged circulation. Moreover, liposomes with a positive charge tend to exhibit relatively toxicity levels compared to neutral or negatively charged liposomes. 168. Aside from their short circulation half-life, unacceptable toxicity, and high net-positive charge, their tendency to aggregate with erythrocytes is the primary limitation to the clinical translation of cLNPs 169. The concept of using ionizable cationic lipids can be a solution to (i) the low-transfection efficiency of LNPs comprised of uncharged lipids and (ii) their biocompatibility issues.
Ionizable Cationic LNPs (iLNPs)
The quaternary ammonium head group of cationic lipids was substituted with a titratable moiety to make ionizable lipids. These ionizable lipids have an electrostatic charge on the head group that is pH-dependent on the head group pKa of the lipid. The ionizable containing LNPs are among the most advanced non-viral-based delivery platforms for efficient gene therapy, as they can self-assemble into NPs when mixed with polyanionic nucleic acids. Also, these ionizable lipids, with modifiable pKa values below pH 7 (at a pH less than 6) are positively charged, thereby allowing high encapsulation efficiency (EE%) of negatively charged nucleic acids such as mRNA, siRNA. When injected, the LNPs have a neutral charge in physiological environments with a pH above their pKa, allowing them to evade uptake by the RES, increase circulation and decrease toxicity associated with positive charge 162. However, once LNPs are taken into the endosome, where the pH is lower than the pKa of ionizable lipid, the amine group of the ionizable lipid loses a proton and binds to the negatively charged endosome lipid head groups. This interaction facilitates endosomal escape 170. Liu et al. described the safe use of aminoionized LNPs for Cas9 mRNA delivery. In their study, they found approximately 70% of gene editing that caused tumor cell apoptosis. They also observed about 50% of tumor growth inhibition and improved the survival rate by 30% with a single intracerebral injection of CRISPR-LNPs against PLK1. However, when these ionized LNPs were engineered with an antibody targeted delivery, they afforded selective uptake in ovarian tumors, enabled up to 80% gene editing; and increased the survival rate to 80%. The ability to disrupt gene expression opens new avenues for cancer research and treatment 62.
iLNPs have been widely used for the systemic RNA therapeutic delivery. Previously, various ionizable lipid materials were reported for LNP formulations such as C12–200 (1-,1’-((2-(4-(2-((2-(Bis(2-hydroxydodecyl)amino)ethyl)(2-hydroxydodecyl) amino)ethyl) piperazin-1-yl)ethyl) azanediyl)bis(dodecan-2-ol), UK-E12 (proprietary), cKK-E12 (3,6-bis(4-(bis(2-hydroxy-dodecyl) amino)butyl)piperazine-2,5-dione) and DLin-MC3-DMA (4-(dimethyl-amino)-butanoic acid, (10Z,13Z)-1-(9Z,12Z)-9,12-octadecadien-1-yl-10,13-nonadecadien-1-yl ester). These LNPs showed efficient gene silencing in the liver at the dose level of 0.002 mg of siRNA per kilogram (mg/kg). The mechanism of cellular uptake was well-investigated, and adsorption of serum ApoE (apolipoprotein E) on the surface of LNPs was a major factor facilitating the intracellular delivery of LNPs into hepatocytes through low-density lipoprotein (LDL) receptors. In recent years, the medicinal chemistry-guided structure-activity relationship (SAR) approach has led to the development of various novel ionizable cationic lipids. In conjunction with sophisticated formulation procedures such as the involvement of microfluidic approaches (which can carefully tune the size of the resulting LNPs), these novel lipids have swiftly increased the likelihood of successful therapeutic interventions. Some recent advances are also seen in the “next generation” lipid and lipid-like molecule developments to make the mRNA-LNP delivery even more efficient. These lipoids are derivatives of ionizable lipids that are comprised of tertiary amines, but are modified by the addition of C12–200 and cKK-E12. These modified lipids demonstrate a gene-silencing efficacy that is akin to that of DLin-MC3-DMA. 171,172. Hence, it appears that the use of ionizable lipids in LNPs formulations could be an outside-of-the-box approach for CRISPR-Cas9 delivery that affords efficacious gene silencing or expression of target genes towards or against a variety of infectious diseases.
Helper Phospholipids
Along with cationic lipids, a standard mRNA-LNP (lipoplex) also contains helper lipids that are added to the lipid composition to enhance the stability of LNPs. In the early stages of cLNP development (for their use in gene delivery), DOPE (1,2-dioleoyl-sn-glycerol-3-phosphoethanolamine) was used as a helper lipid 173. DOPE has a small head group, a phosphoethanolamine, and two unsaturated bulky oleoyl chains. This typical structure of DOPE gives it a pronounced cone shape and helps its assembly in the non-bilayer hexagonal phase 158, which enhances the fusion of LNPs with cell membranes. This peculiar property is the reason why DOPE is considered a “fusogenic lipid” 174. However, some studies have indicated that LNPs based on DOPE exhibit reduced colloidal stability. This is attributed to their excessive fusogenic properties, which could potentially result in cytotoxicity. 49. For this reason, currently, phosphatidylcholines (PCs) have replaced DOPE as a helper lipid in mRNA-LNP formulations. PCs are a natural component of the cell membrane. Particularly, more saturated PCs such as DSPC(distearoylphosphatidylcholine) and HSPC(hydrogenated soybean PC), which have a gel-to-liquid crystalline phase transition temperature, allow LNPs to demonstrate high stability in the formulation 175. DSPC and CHOL lipid combinations were used to make stable LNPs for the nucleic acid delivery 176. Replacing DOPE with DSPC having 40% ionizable lipid served to increase in vitro gene-silencing efficacy 177. Other studies demonstrated that a higher molar ratio of zwitterionic phospholipids in an LNP composition can improve its delivery 77,178.
Cholesterol
Cholesterol is a natural cellular component. It is regarded as a membrane “stiffener”, which induces an intermediate state by increasing the fluidity of the lipid hydrocarbons below and decreasing the fluidity above the Tc 179. Once LNPs are internalized into cells, cholesterol converts the lamellar phase of LNPs into a hexagonal phase by lowering the transition temperature. Therefore, cholesterol helps mRNA release into the cytosol from the internalized LNPs 180. Cholesterol also acts as lipid bilayer stabilizer in both liposomes and LNPs by filling in gaps between phospholipids 181. Cholesterol helps to stabilize LNPs from serum protein. It also promotes membrane fusion in vivo, and thus enhances intracellular delivery 176. Increased molar fraction of cholesterol in DC-Chol-DOPE (3β-[N-(N’,N’-dimethylaminoethane) carbamoyl] cholesterol and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine) DNA lipoplexes demonstrates a higher DNA transfection efficiency 182. Results from a different study indicated that the circulation half-life and the pharmacokinetics (PK) of LNPs improved when 30% cholesterol is mixed with pure DSPC. The circulation half-life increased from a few seconds to five hours. However, adding a higher mole fraction of cholesterol did not further enhance the circulation time 183. Some recent studies are investigating natural cholesterol analogues such as betasitosterol’s, fucosterols, campesterol, and stigmasterol as alternatives to cholesterol. Incorporation of analogues instead of cholesterol appears to improve mRNA transfection efficiency 154
PEG Lipids
The careful construction of an LNP formulation having a suitable combination of excipients is the most critical aspect in optimizing pharmacokinetic and pharmacodynamic (PD) parameters. However, in doing so, it is important to curtail potential adverse effects from activation of the immune response. That is, an LNP should demonstrate negligible activation of immune cells during in vivo administration. PEG has been widely used as a coating that offers stealth-like properties 184. The recent development of PEG-lipid was a recognizable milestone for the safe efficacious clinical use of LNPs. The shielding effect by PEG‐lipids protects the LNP surface against opsonins; protects LNPs against uptake by the mononuclear phagocytic system; and prevents their aggregation when in circulation 185. This multifunctional role of shielding lipids is also beneficial towards its production and storage, as it prevents aggregation and affords LNPs the ability to maintain their nano-sized hydrodynamic diameter. The addition of a PEGylated lipid can improve LNPs colloidal stability in vitro and circulation time in vivo. To improve the bioavailability and circulation half-life of these LNPs, PEGylation helps to shield nanoparticles from opsonization and recognition by the immune system, thereby leading to their prolonged systemic circulation. The shielding effect of PEGylation on LNPs depends on the LNPs’ core structure, lipid composition to which PEG is anchored, nanoparticle elasticity, and PEG molecular properties 186 176 187. The “PEG dilemma” (i.e., the sway between its benefits mentioned above and its drawbacks from the decreased uptake that impede endosomal release) can be exploited by leveraging this property to afford its intentional gradual release into the blood circulation. Immunogenicity and toxicity of dynamic-poly-conjugates are usually unanticipated as is the extent of efficient functional delivery. Patients who have ingested PEG-containing items, but not PEGylated drugs, can develop anti-PEG antibodies. As a result, administering PEGylated medications to those person who have developed anti-PEG antibodies can lead to rapid clearance, decreased therapeutic efficacy, hypersensitivity, and even life-threatening reactions 150. pH-Sensitive anionic helper lipids can trigger low pH-induced changes in LNPs surface charge and stabilization, both of which can facilitate the endosomal release of genetic materials 176.
mRNA-LNP Preparation Methodologies
Thin-Film Hydration
Various preparations were implemented to encapsulate genetic material in LNPs. There are several ways to encapsulate electrostatic interactions between the genetic material and cationic lipids. Conventional LNPs methods are based on a lipid film preparation along with subsequent hydration with an aqueous buffer that contains the genetic material, which allows for passive encapsulation of the genetic payload. Thin-film hydration is the most common and simplest method for mRNA-LNP production. These hydrated vesicles with heterogeneous particles (with diameters > 100 nm) with multilamellar vesicles was formed then require undergoing an additional down-sizing procedure, such as their extrusion or sonication. However, this method is difficult to scale-up and lacks extensive reproducibility. In early 2000, ethanol injection and microfluidic techniques were introduced to overcome these large-scale LNP manufacturing challenges 188–191. Initially, lipids are dissolved in an organic solvent (ethanol and chloroform) and subsequently transferred to a round bottom flask. Next, the solvent is removed using negative vacuum pressure, thereby forming a thin lipid film on the inner surface of the round bottom flask. Next, the mRNA is dissolved in an appropriate aqueous buffer. Hydration of the lipid film with this mRNA-containing buffer results in producing mRNA-loaded LNPs. As electrostatic interaction plays a major encapsulation role. pH adjustment (to pH 4) creates an environment that influences the lipid charge allowing for the interaction with anionic genetic materials. Once lipid bilayer vesicles form with RNA/DNA, ethanol and unencapsulated materials are separated (by dialysis or filtration) to stabilize the LNP. As materials come encapsulated, the pH is adjusted to pH 7 to reduce the LNP surface charge so that it becomes biocompatible. The diameter of the LNPs can be selected by passing such through a polycarbonate filter with a specific diameter cut-off pore size. Three types of LNPs are formed by thin-film hydration: (i) small unilamellar vesicles (SUVs), (ii) giant unilamellar vesicles (GUVs), and (iii) multilamellar vesicles (MLVs) 192,193 (Figure 8).
Figure 8.

mRNA-LNP production and purification methods. The figure illustrates the several different techniques used for LNP productions. These include thin-film hydration. This method involves creating a thin film of lipids by dissolving them in a solvent and then evaporating the solvent to leave a thin film of lipids on a surface. The film is then hydrated with a solution containing the molecule to be delivered, and the LNPs are formed by the self-assembly of the lipids around the molecule. Solvent evaporation is a method involves dissolving the lipids and the molecule to be delivered in a solvent, and then slowly evaporating the solvent to allow the LNPs to form. Emulsion-based synthesis is a method that involves the formation of an oil-in-water or water-in-oil emulsion, in which the lipids and the molecule to be delivered are suspended in one of the phases and the other phase is an aqueous solution. The LNPs was formed by the self-assembly of the lipids around the molecule as the solvent is evaporated. Forth, LNPs production by T junction mixture is a method of creating LNPs using a T junction mixture. This method involves creating a mixture of lipids and other ingredients, such as mRNA in a T junction reactor. Last, Microfluidics technology for LNPs production; A method involves the use of specialized microfluidic devices such as PNI to create and manipulate mRNA-LNPs in a controlled manner. There are several methods for purifying mRNA-LNPs including that includes Amicon centrifugation, gel filtration chromatography, size-exclusion chromatography, TFF and dialysis methods are the most common methods for purification of mRNA-LNP (The figure was created internally by the authors).
Solvent Injection and Emulsion Evaporation
Herein, lipids are first solubilized in water-miscible solvents (isopropyl alcohol, ethanol, and acetone). Next, the mixture is introduced into an aqueous solution with or without surfactants by utilizing an injection needle. The presence of surfactant in the aqueous phase supports the formation of lipid droplets at the injection site whilst concurrently stabilizing the LNPs by decreasing the surface tension between the solvent and water. The resultant mixture is filtered through filter paper to remove the excess amount of lipid present 194. This LNP preparation method exhibits several advantages over other preparation methods including fast manufacturing without the use of expensive devices, incorporation of safe and acceptable organic solvent, and their easy handling 195. An additional method that can be used in mRNA-LNPs formation is the emulsion evaporation method. In this method, initially lipids are dissolved in an organic solvent. Next, an aqueous phase (containing mRNA and surfactant) is added to generate an emulsion. Subsequently, the organic solvent is removed by evaporation under negative pressure. Evaporation of the organic solvent leads to the formation of mRNA-loaded LNPs in the aqueous phase 196. This approach includes single-emulsification and double-emulsification methods. If the formulation is an “oil-in-water” or “water-in-oil system”, then it will be a single emulsion formulation. On the other hand, a double emulsion is formed when the formulation contains “oil-in-water-in-oil” or “water-in-oil-in-water” 197 (Figure 8).
Newer LNPs Production Methods
The major drawbacks of the above-mentioned “conventional” methods for preparing LNPs include their lack of scalability and reproducibility. Large-scale preparation of LNPs that have narrow polydisperse particle size(s) and consistent morphology is challenging due to variations in mass-transport processes and varying production conditions. In recent years, microfluidic mixing techniques have been extensively explored for their scalability in GMP synthesis with low polydispersity index (PDI) which helps to develop homogenous LNPs formulations, that ultimately results in higher targeting efficiency and enhanced biodistribution. It is a versatile technique that has been used to encapsulate a variety of macro-biomolecules such as mRNA, plasmid DNA, and gold nanoparticles into LNPs. Furthermore, siRNA encapsulation efficiencies approaching 100% have also been achieved, which has afforded potent in vivo gene silencing 198 (Figure 9). This technology is mainly based on an ethanol-dilution method. For such, the lipid components are dissolved in ethanol to make a lipid solution, with subsequently adding DNA or RNA to a suitable buffer, such as citrate, acetate, and maleic acid buffer. These two solutions are then passed through a microfluidics chip at a specific flow rate ratio (FRR) and a total flow rate (TFR). By the electrostatic interaction between the negatively charged RNA and positively charged cationic lipids, RNA-loaded LNPs are formed. The dilution of ethanol with an appropriate buffer is a mechanism that helps to form the LNPs 65. The flow rate controls the particle size, whereby small monodisperse particles are produced with faster mixing of the aqueous and ethanol solutions. During mixing, the increase in polarity leads to rapid nucleation of nanoparticles and conversion of the intermediate disk-like planar fragments to vesicles, that prevents growth increases in particle size. This microfluidics-based approach provides advantages over other methods. For instance, lipid-based nanoparticle RNA delivery from a laboratory setting to practical applications allows for accurately tuned particle size with high reproducibility 237, 238. Some of the recent advances were also made on T-Junction for production of LNPs. T-Junction mixing enables controlled and reproducible particles which involves rapid and turbulent mixing of two input streams at a T-junction controlled by adjusting the flow rate. Higher flow rates yield smaller particle sizes, while lower flow rates may result in higher PDIs. Flow rate adjustment allows for fine-tuning of particle characteristics 199
Figure 9.

LNP formulation schemes using microfluidics 200,201. All the lipids are dissolved in an organic solvent, whereas the nucleic acid (mRNA or siRNA) is dissolved in aqueous media. Both solutions pass through a chip at a specific flow rate ratio and total flow rate to achieve nucleic acid encapsulated LNPs. In the last decade mixing tools and fabrications of microfluidic devices have allowed for a considerable improvement in the manufacturing of LNPs. Various mixing and microfluidic devices are described in Table 1 (The figure was created internally by the authors).
Characterization
Gene delivery employs amphiphilic lipids, characterized by a hydrophobic tail and a hydrophilic head 206. Typically, an amide linkage facilitates the interaction between the hydrophobic tail and the hydrophilic head, resulting in the formation of amphiphilic lipoids. This interaction is crucial for incorporating protein components within the membrane, as these proteins harbor hydrophobic segments. These hydrophobic segments engage in hydrophobic interactions with the phospholipid tails, contributing to the stability of the proteins. The resulting structure was confirmed by 1H and 13C NMR spectra 62. Physicochemical characteristics of the LNPs, such as size, shape, charge, polydispersity, and surface composition, each play a vital role in the cellular internalization and resultant efficacy of the particles. Interestingly, the size of the LNPs can be optimized to target specific organs 207. Choice of administration route also depends on LNP size in some cases 208,209. Undoubtedly, size significantly influences different parameters. Thus, it is important to know the LNPs physicochemical properties. One common method for particle size, size distribution, and polydispersity measurement is dynamic light scattering (DLS). This characterization method is based on the Brownian motion of particles dispersed in a liquid. It can analyze nanoparticle diameter size distribution in the range of 0.5 to 1000 nm. DLS is an indirect method which calculates the particle shape pattern, which might not always accurately reflect the true particle shape 210. Some important factors to be noted in DLS measurements include sample dilution needed to avoid multiple scattering and particle-particle interaction. This could result in the destabilization of the formulations. Also, the hydrodynamic radius of the particle measures is critical in the evaluation of DLS measurements. This assumes the particles are in the form of spheres. It need be noted that signals from large particles can mask the signals from smaller particles 211. Another method of size distribution measurement, that also happens to be based on Brownian motion of particles, is nanoparticle tracking analysis (NTA). Hydrodynamic diameter is calculated via using the Stokes-Einstein equation. NTA gives more accurate size distribution calculation than DLS. Still, it has similar limitations like DLS such as experiencing the effects of sample dilution 212. Size exclusion chromatography (SEC) can also be used in size distribution measurement. SEC is mainly used to purify the sample by removing free drugs from the drug-loaded NPs. The separation of particles primarily depends on the pore size of the solid or gel matrix 213. LNP morphology can be identified by the microscope technique, such as negative-stain TEM (transmission electron microscopy) and cryo-TEM (cryo-transmission electron microscopy). Cryo-TEM gives detailed information on particle shape, size, and the membrane structure 214. Another important physicochemical parameter is surface charge (i.e., ζ-potential) of the lipid nanoparticles, as such impacts cellular internalization 215. The surface charge of LNPs usually depends on the phospholipid head groups, which incorporates positive-charged amines or negatively-charged carboxylate groups. The ζ-potential of an LNPs formulation indicates particle stability, as it regulates the strength of particle-particle interaction 64. For stable suspensions, a ζ potential of less than −30mV or more than 30mV would provide sufficient inter-particle repulsion 216. One study suggested that the ratio of DOPE and DOTAP was a critical factor affecting the ζ-potential 65. Another study investigators observed that the ζ-potential of LNPs has a linear relationship with the molar ratio of ionic lipids in the lipid composition of an LNP formulation 64. LNPs are physically stable (in terms of maintaining homogenous distribution and preventing self-aggregation) and maintain EE% during storage time. Here, EE% represents the percentage of a prepared drug that is encapsulated within the LNPs. EE% is determined after the LNPs are prepared and purified, ensuring that any free mRNA or mRNA adsorbed on the LNP surface is removed. EE% remains constant during storage serving to represent the initial amount of mRNA loaded into the LNPs. Förster resonance energy transfer (FRET) serves as a fluorescence-based technique permitting assessment of the quantify of LNP fusion 217. This serves to expand prior works which examined the stability of the Cas9:sgRNA/80-O16B, 80-O16B, Cas9:sgRNA/80-N16B, 80-N16B NPs by measuring the change in average hydrodynamic diameter (<Dh>) over time; wherein a lack in Dh change suggested that LNPs maintained high stability and displayed negligible aggregation after two days of storage at room temperature 62. In another study, a novel polyethylene glycol phospholipid-modified cationic lipid nanoparticle (PLNP)-based delivery system was developed then analyzed for transfection stability. For this, the investigators compared the stability of representative PLNP/DNA-a to that of a control analogue (lipofectamine 2000-encapsulated Cas9-sgPLK-1a DNA (Lipo2000/DNA-a), a commercially available reagent) in different culture media with varying serum concentrations. They found that the size of the Lipo2000/DNA-a particles increased with an increase in serum concentration. The results indicated that PLNP/DNA-a was more stable than Lipo2000/DNA-a in an environment having a high serum concentration 65. Similarly, the stability of the PLNP/DNA was evaluated in vitro at different pH conditions. Their studies revealed that lower pH values increased the diameter, thereby highlighting the instability of the particle’s structure. More specifically, these results indicated that PLNP/DNA particles could decompose under acidic conditions (e.g., in the endolysosomal system due its intravesicular pH of ~5) 65. The stability of 5A2-DOT-10 LNP encapsulated with Cas9GFPP RNP complexes was determined throughout storing such at 4°C for two months. The PDI (polydispersity index) and size of the nanoparticles were tested at different times of storage. Analyzing the particle size diameter at different timepoints revealed that the size of the LNPs did not change and remained uniform (PDI < 0.2). Similarly, gene-editing efficiency was evaluated in HeLa-GFP cells after adding 24 nM sgRNA nanoparticles, with gene-editing quantification carried out after 3 days. The gene-editing efficiency of the 5A2-DOT-10 remained constant even after its storage of 60 days. 218. How much mRNA is loaded during LNP preparation and remains encapsulated during storage is crucial in mRNA-LNP development and characterization. The RiboGreen® assay was conducted to determine the EE% of sgRNA and Cas9mRNA in various lipids viz: L6-, L8-, L10-, and MC3-LNP obtained from the lipid library. The EE% was found to be high (> 90%) for the various nanoliposomes except for L1-cLNP (~65%) 206. The CRISPR-Cas9 LNPs were formulated for a single administration for persistent in vivo gene editing. In another study, 61.5% EE% of CRISPR-gold was found for complex Cas9 RNPs, which met the necessary efficiency for developing Cas9 delivery vehicles 65. Zhen and co-workers used an encapsulation ratio (%) of [(A2-A1)/A2] ˣ100 % to calculate the CRISPR-Cas9 crude content in the supernatant. The EE% of the nanoliposome complex was 96.87% ± 0.13 219. The EE% of the complex was further confirmed using gel retardation electrophoresis. The EE% of the complex was determined to be 95% by performing a grayscale analysis 219. Stability should also be measured in terms of gene disruption efficiency. The cLNP formulations encapsulating GFP sgRNA were evaluated for in vitro gene disruption efficiency by analyzing GFP-associated fluorescence signal loss (via monitoring reduction of GFP signal) in HEK 293 cells expressing GFP (HEK293/GFP) for cancer therapy 206. In all, LNPs should be rationally designed such that they retain the mRNA payload during storage as well as efficiently release. The CRISPR-Cas9 gRNA-HPV16 E6/E7 nanoliposome complex release profile was evaluated after fitting the composite object release model as per the release profile in vitro. The R2 value was found to be higher in the Higuchi equation, thereby indicating that the nanoliposome complex conformed to the nanoliposome characteristics. Also, the complex release rate was 37.7% at 3 hours. This meets the required liposome formulation guidelines as per the relevant pharmacopeia 219. mRNA expression following its release after being taken up by its targeted cells can be measured using flow cytometry. The expression level of GFP and its congener BFP (blue fluorescent protein) in the CRISPR-gold treated BFP-HEK was quantified using flow cytometry. The analysis of BFP-HEK cells was performed after seven days of Cas9 treatment. The level of GFP and BFP expression was quantified from the visualized fluorescence intensities from localized cell specimens using fluorescence microscopy 65.
Routes of LNPs Administration
In vivo biodistribution (BD) and PK of LNPs are highly affected by the selected route of administration 220. Moreover, its influence on the strength of the immune response as well as any potential side effect(s) makes it an important point of consideration. LNPs serve as unique delivery vehicles for transfection of genetic material that offer the ability to target specific cells and effectively deliver the therapeutic cargo to relevant intracellular sites. In the past few years, various delivery routes have been explored for mRNA-LNP delivery.
Parenteral
The first demonstration of parenteral delivery of LNPs surfaced in 1960 with the use of a parenteral fat emulsion of submicron size for parenteral nutrition (Intralipid®221. Intravenous (IV) administration is preferably chosen when it is desired for mRNA-LNPs to be delivered into the liver. The major concerns with this delivery route were the extent of systemic toxicity and immunogenic response 222. Therefore, IV delivery of mRNA-LNP is uncommon. Other parenteral routes include intramuscular (IM), intradermal (ID), and subcutaneous (SC) routes of administration (Figure 10). IM route remains the most common form of delivery and is easy to carry out. IM delivery of mRNA using LNPs has shown successful experimental outcomes in many preclinical and clinical studies 171. For instance, a robust immune response was observed by IM delivery of mRNA LNPs vaccines against H10N8 and H7N9 influenza viruses. This was seen in a phase 1 clinical trial 223. Intradermal (ID) injection involves the administration of LNPs directly into the dermis, directly below the epidermis. It was observed in some studies that ID administration requires one-fifth of a standard IM dose to achieve an equivalent response. 224,225 In another study, mRNA-LNP vaccines administered via the ID route effectively induced a Th1 immune response and efficiently primed cytotoxic T-cells. Subcutaneous (SC) delivery for mRNA-LNP administration permits the LNPs to be administered to the connective tissue. Noteworthy, SC administration of mRNA-LNPs encoding fibroblast growth factor 21 (FGF21) was performed. This was for use of a therapeutic agent in type 2 diabetes (T2D) and non-alcoholic steatohepatitis (NASH). A therapeutic level of FGF21 following such administration was observed 226.
Figure 10.

The route of administration of the delivery vehicle affects the BD and PK properties of the desired cargo (The figure was created internally by the authors).
Local Administration
Several in vivo studies involving local administration of CRISPR-Cas9 with LNPs have shown promising results, such as their direct delivery in the inner ear 227, muscles 65, brain 22,228, and tumors 229. Localized ear delivery of Cas9:guide RNA complexes with cationic lipid mediated LNPs ameliorated hearing loss in a model of human deafness. They have designed (and validated in primary fibroblasts) in vitro genome-editing agents that preferentially disrupt the dominant deafness-associated allele in the Tmc1 (transmembrane channel-like 1) Beethoven (Bth) mouse model. Then, they subsequently injected Cas9:guide RNA lipid complexes that targeted the Bth allele into the cochlea of neonatal Bth/+ mice. They found substantially-reduced progressive hearing loss, higher hair cell survival rates, and lower auditory brainstem response (ABR) thresholds in injected ears compared to ears not injected with such or ears injected with complexes that target an unrelated gene 227. In another study by Lee et al, they demonstrated localized muscle delivery of vehicles composed of gold nanoparticles conjugated to DNA and complexed with cationic endosomal-disruptive polymers. This multi-complex NP was used to transport the Cas9 ribonucleoprotein and donor DNA following local injection such that efficient correction to the DNA mutation that causes Duchenne muscular dystrophy transpired with minimal off-target DNA damage 65. Wang et al. reported tumor-localized delivery of the Cas9 protein and sgRNA plasmid using gold nanoclusters (GNs) encapsulated in the core of the nanocapsules. They modified the GNs with HIV-1-transactivator of transcription peptide, the cargo (Cas9/sgRNA) that can be delivered into cell nuclei that is subsequently utilized to treat the melanoma by designing sgRNA targeting Polo-like kinase-1 (Plk1) of the tumor. This nanocapsule (polyethylene glycol-lipid/GNs/Cas9 protein/sgPlk1 plasmid, LGCP) led to a > 70% down-regulation of Plk1 protein expression in A375 cells in vitro and suppressed melanoma progression by 75% in mouse models 229.
Inhalation
Direct delivery of mRNA to the lungs can be achieved through nebulization. Nebulized mRNA LNPs show distinct delivery outcomes compared to systemically administered LNPs. The biological environment in the lungs during nebulization differs in terms of biomolecules and cell types, presenting unique barriers for LNPs compared to bloodstream administration 230. Moreover, aerosol droplets experience shear stress, which causes particle fragmentation and decreases particle stability 231. Some studies have demonstrated that lipid composition optimization can address this problem. For example, a higher molar concentration of PEGylated lipids was found to enhance the efficacy of mRNA LNPs administered by nebulization. They found that optimized mRNA-encoded LNPs protected mice from the H1N1 subtype of the influenza A virus more efficiently than the optimized mRNA LNPs administered systemically 230.
Intranasal
The conventional mRNA LNPs administration methods that include intravenous, IM, ID, and subcutaneous routes are associated with injection-related effects (pain, trouble with repeated administration, risk of infection, and low patient compliance). The intranasal route for mRNA LNPs delivery is one of the more ideal noninvasive routes of administration, as administration can be repeatedly performed with low infection risk and high patient compliance. In addition, higher permeability and vascularization of nasal mucosa make the nasal tissues more accessible than other tissues 232,233. It is evidenced that mRNA LNPs can be given by nasal administration. Due to their colloidal nature, LNPs can be readily transported and distributed via their drainage throughout the lymphatics within the nasal cavity. Brain delivery is also benefited from this route of administration since LNPs can be delivered directly to the olfactory lobe, thereby bypassing the blood-brain barrier (BBB) to allow for higher bioavailability 234,235. Several studies have demonstrated more promising results for mRNA LNP delivery to the brain via utilizing the intranasal route of delivery 236.
In utero
Most congenital disorders that occur due to protein deficiencies are treated with postnatal protein or enzyme supplements. We believe the high morbidity and mortality rates associated with these irreversible disorders can be minimized with the use of prenatal in utero mRNA delivery. Although viral-based vectors have been explored for this purpose, their higher immunogenicity and low genomic integration limit their application 120. Several studies have been performed to optimize the efficacy of LNPs by screening from a library of LNP formulations for effective in utero mRNA delivery to the fetal liver, lung, and spleen. A wide range of mRNA including GFP (green fluorescence protein) and EPO (erythropoietin) mRNA has been found to be efficiently delivered by using a library-optimized LNP formulation 237. Library design is based on the advantages seen through recent developments of mRNA-LNP vaccines. These were produced based on a developed platform and the rapid manufacturing. The screened libraries led to an optimal mRNA-LNP vaccine for efficacy, stability, and toxicity. Each in composite highlights the critical aspects uncovered in the design of mRNA-LNP platforms that include the means to optimize those platforms.
Ocular Administration
Gene therapy via utilizing ocular administration is advantageous due to several reasons, such as affording a relatively low immune response and being readily effectuated. The use of a viral-based vector can be an option for ocular mRNA delivery; however, the major concern lies in its immunogenic response and potential for long-term toxicity 238. Non-viral-based vectors, especially LNPs, are found to be an attractive alternative to viral-based vectors due to their low immunogenicity and high safety profile 5.
Administration Routes
As mRNA-LNPs can be delivered through many different routes, their biodistribution, expression kinetics, and therapeutic efficacy is highly influenced by such 220. The specific application or specific desired outcome largely governs the route choice for the delivery of mRNA LNPs. For example, in the case of genetic disorders related to protein deficiency, mRNA LNPs delivery via utilizing IV can be considered a viable option for its administration route. The reasoning behind this is that when mRNA LNPs are administered intravenously, they primarily accumulate in the liver. As the liver is an organ that inherently produces an array of proteins, their accumulation in such lends itself the potential to affording improved outcomes. That is, IV administration can be used to produce proteins that are deficient in congenital, metabolic, or hematologic disorders 239. However, the major drawback of their IV administration is their tendency to activate the immune response and undergo high systemic clearance 240. Also, it can lead to unwanted accumulation of LNPs in different organs and induce off-target side effects. As such, the introduction of LNPs via performing IM, ID and SC injection are preferable routes of administration due to predefined tissue-specific delivery. The results from several studies have indicated that local administration of mRNA LNPs can effectively afford the expression of tissue-specific protein supplements, such as those associated with the eye, brain, and heart 238,241,242. For treating prenatal congenital diseases, the delivery of LNPs via in utero administration can be the option of choice for achieving protein expression in the fetal liver, lung, and spleen 120,237. Again, repeated drug delivery by invasive methods is not feasible in these instances. In such cases, inhalation and the intranasal route of administration can make mRNA delivery more patient-friendly. Additionally, the intranasal route can give better results when the mRNA LNPs are intended for brain delivery 235.
Targeted LNPs Delivery
Targeted mRNA LNP delivery can be a promising means to increase therapeutic efficacy and mitigate potential off-target effects by nontargeted tissue. One approach to make the LNPs target-specific is called selective organ targeting (SORT) 49. To create SORT the investigators added a fifth lipid along with the four standard lipids for mRNA LNP formulations, which are ionizable cationic lipids, helper phospholipids, cholesterol, and PEGylated lipids. They modified the lipid composition based on the notion that the internal and external charge of LNPs affects their biodistribution 243. They observed that adding a permanently charged 1,2-dioleoyl-3-trimethylammonium-propane lipid (DOTAP) at an increasing molar percentage (0 to 100%) shifted luciferase expression from the liver to the spleen to the lung. Incorporation of a negatively charged SORT molecule, 2-dioleoylglycerolcero-3-phosphate at 10 to 40% achieved spleen-specific luciferase expression. They also observed that by using several other classes of permanent and ionizable lipids, they attained similar outcomes, thereby bolstering the utility of their approach. As an extension of their efforts, the same group explored the use of helper phospholipids. For doing so, they synthesized ionizable phospholipids and optimized them in a manner that afforded tissue-specific mRNA delivery as well as improvements in endosomal escape of mRNA 21. Currently. cancer is the primary cause of death. The major concern with conventional treatments (traditional chemotherapy) is the effect of off-target drug delivery. Intratumor LNP injection may afford promising results. However, it is more invasive. To overcome this barrier, a noninvasive anchored secondary scFv approach was developed. This method involves a targeting antibody that is added to the LNP membrane 244. The pKa value of the lipids, which affects the ionization status of the LNP membrane, may also influence the scFv-based tissue-specific delivery by altering the electrostatic interactions with specific tissue. In one study, researchers found that LNPs having lipids with lower pKa values on the membrane, such as 6.45 or 6.5, tend to distribute throughout the hepatocytes of the liver, whereas LNPs that have membrane lipids with higher pKa values, such as 6.8 or 7.1, are more likely to be routed to the liver sinusoidal endothelial cells (LSECs) 245. Another prospect for achieving targeted tissue-specific delivery would be the specific modified mRNA translation systems (SMRTs). This is an approach that entails a self-controlled expression of mRNA inside a specific type of tissue irrespective of its cellular uptake by other tissue types. It is an on-off system containing two mRNA together in a single circuit (in series), which controls the mRNA expression in select tissue types and prevents expression in non-targeted tissue 246. Conjugating CD4+ antibodies to LNPs was found to lead to specific targeted and efficient mRNA translation in CD4+ T-cell populations demonstrating the efficacy of mRNA-based immunotherapeutic delivery. The study demonstrated that in vitro targeting of human T-cells with anti-CD4+-conjugated luciferase mRNA, resulted in robust binding (measured by radioactivity and flow cytometry) and efficient luciferase expression in CD4+ T-cells. They also assessed the functional potential of the conjugated mRNA-LNPs to facilitate genome editing using the Cre-LoxP mice reporter system and demonstrated successful genetic recombination (~60% efficiency in spleen), irrespective of the activation status (resting or activated) of CD4+ T-cells. Furthermore, a timepoint analysis showed a gradual decrease in the expression of mRNA in the spleen compared to the stable expression in lymph nodes. They suggested that a plausible reason for such could be the natively longer residence time of T-cells in lymph nodes than in the spleen. In sum, the study presented a CD4+ T-cell-targeted LNP platform for in vivo delivery of mRNA therapeutics 247. Most recent study by Verve Therapeutics’s team developed an optimized GalNAc-LNPs for targeted CRISPR base editing therapy in patients with low-density lipoprotein receptor (LDLR) deficiency by optimized N-acetylgalactosamine (GalNAc) for asialoglycoprotein receptor, ASGPR)-based ligand enhanced liver editing while reducing off-target effects. Their GalNAc-LNPs demonstrated high editing efficiency in LDLR deficient non-human primates and achieved long-lasting protein reduction in wild-type monkeys (https://www.nature.com/articles/s41467-023-37465-1)
PK and BD
Systemic delivery of gene therapy systems often results in rapid degradation and/or prompt clearance of the material. The genetic material can be protected from degradation by encapsulating it within a stable non-viral-based vector (e.g., in LNPs). One of the primary reasons attributed to the success of this approach is that LNPs can achieve better biodistribution and pharmacokinetic profiles of gene delivery in vivo 248. Replacing the hydrogen or carbon atoms of nucleic acids with radioactive isotopes 3H or 14C has little effect on the structure or PK behavior of the nucleic acid 249. However, van de Water et al. 248 found that the biodistribution and pharmacokinetics are different when they used 3H-versus 111In-labeled unformatted siRNA. Hence, it is essential to choose a suitable isotope to monitor the PK and biodistribution behavior of LNPs. Studies used fluorescence imaging, due to its low cost and relative safety, to monitor the distribution and pharmacokinetics of LNPs during gene therapy 49,103. One report explained that the LNP-gene therapy having positive charge could bind to different types of serum proteins, including heparin, albumin, and lipoprotein 250. The binding strength between the two is influenced by the charge density and surface structure of the lipid/plasmid complex. Also, extensive lung accumulation was observed likely due to entrapment of the LNP-gene formulation in lung capillaries 251. The build-up of these in the lungs may be caused by the ionic interaction with the extensive surface area of endothelial cells. However, others found that negatively-charged LNP-gene complexes do not lead to lung accumulation 252. When DOPE and a stearylated octa-arginine (STR-R8) peptide were employed as the inner coat and the outer coat consisting of cationic lipid di-octadecyltrimethylammonium propane-cholesterol-YSK05 distribution can be amended. In this instance optimized amounts of YSK05 (1-methyl-4,4-bis[(9Z,12Z)-octadeca-9,12-dienoxy]piperidine) and GALA (glutamic acid-alanine-leucine-alanine) peptides were used, which produced a high gene expression level in lung (>107 RLU/mg protein) with high lung:liver and lung:spleen ratios. The investigators used the same exact amount and type of lipid and the same pDNA dose in a 1-step/2-step coating. The results showed that transfection activity was higher in lungs in the case of 2-step coating. In all double-coating improved the efficiency of gene expression.
Conclusions
Viral vectors are commonly used for CRISPR-Cas9 delivery, but they have drawbacks like insertional mutagenesis, undesired tissue expression, immune reactions, and translational challenges. Non-viral alternatives offer advantages such as biocompatibility, low biodegradability, minimal immunogenicity, tissue targeting, and the ability to deliver various payloads. These non-viral vectors aim to overcome limitations of viral vectors, including low loading efficiency and the need to split CRISPR-Cas9 components, to achieve effective delivery of CRISPR-Cas9 systems ex vivo and in vivo. 229,253,254. Non-viral vectors offer advantages such as scalability, reproducibility, and cost-effectiveness over viral vectors, making them a preferred drug delivery option. However, addressing challenges like lower editing efficiency and payload issues requires further research. Recent advancements in nanotechnology have led to the exploration of various non-viral delivery systems for genome editing. Lipid-based synthetic nanoparticles/liposomes, polymeric/ polypeptide-based carriers, and natural extracellular vesicles are among the well-studied systems for CRISPR-Cas9 delivery 206. The development of non-viral-based vectors and lipid-based systems for delivering CRISPR cargos holds promise for in vivo gene excision. Factors affecting CRISPR-Cas9 editing efficiency include sgRNA sequence, nuclease exposure duration, delivery efficiency, and DSB repair mechanism. Design factors for nanoformulations also influence in vivo gene editing proficiency. However, developing delivery systems for clinical acceptance remains challenging. Selection of appropriate components for lipid-based LNPs depends on cargo carrier characteristics, lipid interaction, physicochemical properties, and tissue-targeting potential. Variations in gene disruption correlations raise questions about non-viral-based systems. Understanding CRISPR cargo carriers and lipid roles is crucial for LNP development. Effective delivery for therapeutic efficacy against viruses like HIV requires avoiding liver metabolism/clearance and targeting spleen and lymph nodes. Understanding each step of non-viral lipid-based vector gene transfer is necessary. Critical parameters for nanocomplex formulations include particle size, ζ-potential, charge ratio (N/P), endosomal escape, gene-loading efficiency, and release profile. Optimal electrostatic interactions between nucleic acids and lipids are important for encapsulation and binding. Various cationic lipids are widely used as nonviral-based gene carriers due to their high transfection efficiency 229,255. Optimizing the ratio of cationic and neutral lipids to gene payload is crucial. Lipoplexes have demonstrated great potential for in vitro, ex vivo, and direct delivery of Cas9 plasmid/mRNA/protein and sgRNA. However, effective delivery of cargo carriers remains a challenge for CRISPR-based human engineering. Escape from the endosomal pathway by peptides (paradexin) 256, PEI-mediated osmotic pressure and liposome-mediated membrane fusion 257, use of pH-responsive swelling of lipids/polymers, phototherapy-based approaches 62,258, NIR irradiation 49 and cationic peptide-mediated membrane destabilization. All were explored in attempts to expanding LNP suitability 206,259. Poor stability of LNPs in biological fluids upon exposure with serum also poses a challenge in the effective delivery of genome editing components 235,260,261. Structural modifications of LNPs to lipid nano-shells offer an alternative cargo carrier system in such cases. These lipid nano-shells can also be explored for multicomponent systems with the Cas9 protein, plasmid DNA containing target genes, gold nanoparticles 229 and/or lipofectamine 65. The targeting potential of LNPs also remains a critical point of consideration, as unwanted expression or undesirable tissue site targeting may lead to adverse effects. The risks of off-target cleavage by specificity, which leads to longer exposure to the Cas nuclease could result in undesired mutation 37,38,262–264. Mismatch of sgRNA, due to tolerance of the Cas9 nuclease, results in expression at unsought genomic locales. Such undesirable genome editing in adjacent tissues also results in such adverse reaction and low therapeutic spectrum 264. In the recent past, advanced strategies, such as use of double Cas9 nickase 73,264–266, have been explored to reduce off-target expression-associated side effects. Appropriate selection of suitable lipids, polymers and ligand-mediated surface decoration on nano-formulations also offered an alternative strategy for optimizing the expression at target sites. In a recent study, LNPs were explored for the successful delivery of Cas9 mRNA and sgRNA effectively to the liver in order to modulate expression of transthyretin in mouse with 97% reduction in serum protein level even after several months of treatment 163. Recently, SORT NPs for tissue-specific mRNA delivery and CRISPR-Cas9 gene delivery have been explored. In this strategy, re-engineered LNPs by employing supplemental SORT molecule(s) for the targeting of Cas9 mRNA/sg RNA, were devised for selective targeting of lung, liver, and spleen 49. Based on the current understanding of cellular barriers, the molecular structure of cell membrane, and cell surface topology, considerable progress has been made in utilizing ligand-based LNPs for effective targeting that results in limited adverse events linked to reduced off-target expression. RGD-mediated integrin receptor targeting via lipid nano-shells comprised of liposomal templated hydrogels can also improve the genome-editing efficacy of CRISPR-Cas9 components, including sgRNA targeting PLK1 65. Environment-based selective targeting also provides an option by using pH-sensitive lipids that respond to the pH of tumor tissue and endosomes for triggering and open stimulation of delivery systems 267,268. pH-sensitive liposomes were also designed by mimicking the viral fusion with endosomal membrane using a protein at pH 5–6, which allowed for the delivery of their genetic material to the cytosol before reaching the lysosomes 269,270. PE, a lipid-based membrane fusion promoter, is used in such nano-formulations 271. Exploring various cellular markers, and unique receptor epitopes and understanding of physical conditions surrounding the target cells would be ideal. However, phenotypic heterogeneity among targeted cells remains a greater challenge for effective delivery through these approaches. Therefore, LNP-mediated direct delivery of Cas9 protein for rapid action and high efficiency should be further explored in the future to address the problem of off-targeted adverse events. Multi-layered NPs offer an exciting option for delivering CRISPR cargo carriers with considerable in vivo expression efficiency 272. Multi-components such as a cationic core, organic shell and multifunctional ligands on the external surface of these artificial multi-layered LNPs exhibit excellent site-specific delivery, and thus provide improved genome-editing efficiency with low adverse effects 272. For designing therapeutically active CRISPR-Cas9 delivery systems, proper consideration of the route of administration is also warranted to control the in vivo fate of these cargos. The success of IV-administered nano-formulations of CRISPR-Cas9/sgRNA in mouse models appears promising. However, detailed studies on their deposition in organs like liver, kidney, and lungs, as well as on-site expression efficiency, are necessary for effective nano-carrier development. Intra-tumoral administration has demonstrated better safety and site-specific targeting, resulting in successful gene knockout and greater localized effects. 273,274. Ocular delivery or retinal targeting through intravitreal administration routes has also been reported to “knock-in” therapeutic genes using Cas9/PAMAM polymeric NPs 219. Localized infusion of lipid-coated polymeric NP-mediated Cas13a/crRNA into the bladder cavity also showed excellent targeting potential with low systemic adverse effects 206. In view of these studies, safe gene editing at disease-specific targeted organs seems possible due to overcoming the associated challenges of formulation design and selection of compatible components in diseases like retinal degeneration, urinary tract disorders, and a variety of cancers. Furthermore, being of bacterial origin, the risk of SpCas9 in inducing immunogenic reactions in the recipient host is very high due to prolonged expression of bacterial Cas9 protein on MHC-I molecule, thereby resulting in the generation of antibodies and elicitation of T-lymphocyte-mediated immune response 206,275. This immune cascade can result in adverse events, even leading to treatment failure. Designing LNPs faces a challenge due to the self-immunogenic potential of Cas9. While the lipids used in nano delivery systems are biocompatible and non-immunogenic, preventing premature exposure of the encapsulated gene in the system before reaching the target site is crucial to avoid unwanted immune responses. Ensuring effective loading and biochemical modification is essential for designing successful LNPs. Monitoring and evaluating humoral and cellular immune responses during clinical trials, along with considering ease of administration, are important factors that govern efficacy. Interactions with blood components and the inhibitory effects of serum present significant challenges for effective LNP delivery 276. To achieve successful clinical translation of LNPs, designing customized nano-carriers is crucial. This requires a deeper understanding of nano-bio interactions, safety considerations, loading capacity, compatibility with multifaceted strategies, spatiotemporal release, surface stabilization, electrostatic properties, and route of administration. Modifiable LNP components such as size, shape, charge, and surface properties should be tailored for accurate application in specific patient populations.
Table 1.
| Type of mixing device | Staggered herringbone micromixer | Toroidal mixer | T-junction | Hydrodynamic flow focusing | Baffle mixer |
|---|---|---|---|---|---|
| Schematic Design |
|
|
|
|
|
| Advantages | -Controlled mixing -High EE% (>95%), -Uniform particles (PDI < 0.1) -Scalable between small, large batches on parallelization. |
-LNPs encapsulating siRNA by mixing preformed vesicles with siRNA in ethanol- water solutions and by mixing lipids in ethanol aqueous components containing siRNA | -Controlled rapid mixing -High EE% -Uniform particle size, -Broad solvent compatibility |
-Can accelerate lipid convection to the mixing interface by reducing the migration distance of the organic stream -Precise control of LNP size -Low PDI203 |
-High EE% (>90%) - LNP size can be fine-tuned |
| Flow rate capacity | 1–20 mL/min | 1 mL/h to ≥ 20 mL/h | 40–60 mL/min | 0.025–2000 mL/min | 0.1 mL/min |
| Particle size (diameter) | < 100 nm | < 100 nm | < 40 nm | < 150 nm | < 100 nm |
| Scalability | Multiple staggered herringbone micromixers in parallel station is required to achieve scalability under GMP conditions | One chip required to achieve scal-ability under GMP conditions | A general method of choice for the commercial production of large batches of LNPs | Fully scalable manufacturing is possible | Fully scalable manufacturing is possible |
Table 2.
Prototypical clinical trials of LNP-based nucleic acid delivery systems.
| LNP-based nucleic acid delivery systems for cancer therapy | |||||||
|---|---|---|---|---|---|---|---|
| Clinical Trials | Phase | Disease | Interventions | Route | Sponsor | ||
| NCT00882180 | Phase I | Solid tumors | siRNA against VSP/VEGF-A | Intravenous infusion | Alnylam Pharma | ||
| NCT01262235 | Phase I/II | Gastrointestinal Neuroendocrine Tumors | siRNA against PLK1 | Intravenous infusion | Arbutus Biopharma | ||
| NCT02191878 | Phase I/II | Hepatocellular carcinoma | siRNA against PLK1 | Intravenous infusion | Arbutus Biopharma | ||
| NCT03323398 | Phase I/II | Relapsed/Refrac tory Solid Tumor Malignancies or Lymphoma | mRNA-2416 encoding OX40L, alone (Phase I) or in combination with i.v. PD- L1 inhibitor, Durvalumab (Phase 2) | Intratumoral | Moderna TX, Inc. | ||
| NCT03323398 | Phase I | Advanced/metas tatic solid tumors or lymphoma | mRNA encoding human OX40L | Intratumoral | Moderna TX, Inc., | ||
| NCT03313778 | Phase I | Solid tumors | mRNA encoding for personalized neoantigens | Intramuscul ar | ModernaT X, Inc., | ||
| NCT02340156 | Phase II | Recurrent glioblastoma | Plasmid vector encoding wild- type p53 DNA (+temozolomi de) | Intravenous infusion | SynerGene | ||
| NCT01505153 | Phase I | Advanced and/or metastatic Cancer | Plasmid vector encoding shRNAs against STMN1 | Intratumoral | Strike Bio | ||
| NCT02716012 | Phase I | Advanced liver cancer | small activating RNA to activate the CEPBA gene | Intravenous infusion | Mina Alpha | ||
| NCT02410733 | Phase I | Melanoma | Four mRNAs encoding melanoma- associated antigens (MAAs) | Intravenous infusion | BioNTech | ||
| NCT02316457 | Phase I | Triple negative breast cancer | mRNAs encoding tumour- associated antigens (TAAs) and/or personalized neoantigens | Intravenous infusion | BioNTech | ||
| NCT01808638 | Phase Ib/Iia | Advanced or metastatic pancreatic cancer | siRNA against PKN3 (+ gemcitabin e) | Intravenous infusion | Silence Therapeuti cs GmbH | ||
| NCT03739931 | Phase I | Metastatic disease | mRNA-2752- OX40L, IL-23, and IL-36γ mRNA with LNPs and Durvalumab | Intratumoral | Moderna TX, Inc | ||
| NCT01591356 | Phase I | Advanced cancers | siRNA against EphA2 | Intravenous infusion | M.D. Anderson Cancer Center | ||
| LNP-based nucleic acid therapies for systemic diseases | |||||||
| Clinical Trials | Phase | Disease | Interventions | Route of immunizati on | Sponsor (s) | ||
| NCT04601051 | Phase I | Familial Amyloid Polyneuropathy, Transthyretin- Related Familial Amyloid Cardio- myopathy, Wild- Type Transthyretin Cardiac Amyloidosis | NTLA-2001- CRISPR-Cas9 system delivers by LNPs | intravenous | Intellia | ||
| NCT05398029 | Phase1b | CRISPR base-editing medicine to PCSK9 gene turn off | VERVE-101 | intravenous | Verve | ||
| hepatic protein production for heart disease | |||||||
| NCT03767270 | Phase I/2 | Ornithine Transcarbamylase Deficiency | MRT5201-human ornithine transcarbamyl-ase mRNA with lipid- based nanoparticles | intravenous | Translate Bio, Inc. | ||
| NCT04159103 | Phase I/2 | Propionic acidaemia | mRNA-3927 encoding Propionyl- CoA carboxylase | intravenous | Moderna TX, Inc. | ||
| NCT03375047 | Phase I/2 | Cystic fibrosis | MRT5005 | Inhalation | Translate Bio, Inc. | ||
LNPs are a successful delivery vehicle that allow for improved treatment outcomes for vaccines and therapies. This includes treatment and prevention for a broad range of genetic, infectious, and cancerous diseases. Clinical trials for cytotoxic chemotherapy agents, antibiotics, and nucleic acid therapeutics are outlined in this table. LNP-based COVID-19 mRNA vaccines from Pfizer/BioNTech and Moderna were successfully developed with international impact.
Statement of significance.
CRISPR-Cas9 gene editing for disease treatment and prevention is an emerging field that can change the outcome of many chronic debilitating disorders.
Acknowledgments
The authors would like to express gratitude towards Tom Barger and Nicholas Conoan of the Electron Microscopy Core Facility (EMCF) at the University of Nebraska Medical Center for technical assistance. The EMCF is supported by state funds from the Nebraska Research Initiative and the University of Nebraska Foundation and institutionally by the Office of the Vice Chancellor for Research; Victoria Smith and Holly Britton of the UNMC Flow Cytometry Research Facility. The UNMC Flow Cytometry Research Facility is administered through the Office of the Vice Chancellor for Research and supported by state funds from the Fred and Pamela Buffett Cancer Center’s National Cancer Institute Cancer Support Grant. Some of the Figure panels were made with BioRender.com. This publication’s contents are the sole responsibility of the authors and do not represent the official views of the funding agencies.
Funding sources.
This work was supported by NIMH (R01 MH121402–01A1), NINDS (R01 NS034239–28), NIDA (P01 DA028555–06A1), and UNMC student assistantships.
Abbreviations
- AAT
Alpha-1-antitrypsin
- AAVs
Adeno-associated viruses
- AAVs
Adeno-associated-based Viruses
- ABEs
Adenine base editors
- ABR
Auditory brainstem response
- Ad
Adenoviruses
- AIDS
Acquired immunodeficiency syndrome
- AMD
Age-related macular degeneration
- ASSET
Anchored secondary scFv enabling targeting
- AVs
Adenovirus-based Vectors
- BBB
Blood-brain barrier
- Bes
Base Editors
- BFP
Blue fluorescent protein
- BrdU
5-bromo-2′-deoxyuridine
- Bth
Beethoven
- CAR-T
Chimeric Antigen Receptor-T
- CBEs
Cytosine base editors
- CFTR
Cystic fibrosis transmembrane regulator
- cLNPs
Cationic Lipid Nanoparticles
- CReVIS-seq
CRISPR -enhanced viral integration site sequencing
- cryo-TEM
cryo-transmission electron microscopy
- CTL
Cytotoxic T lymphocytes
- DLS
Dynamic light scattering
- DSB
Double-stranded break
- EE%
Encapsulation efficiency
- EPO
Erythropoietin
- FGF21
Fibroblast growth factor 21
- FMLV
Friend Murine Leukemia Virus
- FokI
A type II S restriction enzyme from Flavobacterium okeanokoites
- FRET
Förster resonance energy transfer
- FRR
Flow rate ratio
- GALA
Glutamic acid-alanine-leucine-alanine
- GFP
Green fluorescence protein
- GNs
Gold nanoclusters
- GUIDE-seq
Genome-wide unbiased identification of DSBs enabled by sequencing
- GUVs
Giant unilamellar vesicles
- HCAds
High capacity adenoviral vectors
- HDR
Homology-directed repair
- HIV
Human immunodeficiency virus
- HPV16
Human papillomavirus 16
- ID
Intradermal
- IFN
Interferon
- iLNPs
Ionizable Cationic LNPs
- IM
Intramuscular
- iNPs
Inorganic nanoparticles
- ITR
Inverted Terminal Repeat
- IV
Intravenous
- IVT
in vitro transcription
- LAM-HTGTS
Linear amplification-mediated high-throughput genome-wide translocation sequencing
- Lipo2000/DNA-a
Lipofectamine 2000-encapsulated Cas9-sgPLK-1a DNA
- LNAs
Locked nucleic acids
- LNPs
Lipid nanoparticles
- LSECs
Liver sinusoidal endothelial cells
- LTR
5’- and 3’-Long Terminal Repeat
- mAbs
Monoclonal antibodies
- MHC-I
Major histocompatibility complex-I
- MLVs
Multilamellar vesicles
- MpAgo
Marinitoga piezophila argonaute
- MRSA
Methicillin-resistant staphylococcus aureus
- Nabs
Neutralizing antibodies
- NASH
Nonalcoholic steatohepatitis
- NASH
Non-alcoholic steatohepatitis
- NHEJ
non-homologous end joining
- NPs
Nanoparticles
- NTA
Nanoparticle tracking analysis
- ORF
Open Reading Frame
- OTCD
Ornithine transcarbamylase deficiency
- PAM
Protospacer adjacent motif
- PDI
Polydispersity index
- pDNA
Plasmid DNA
- PE
Prime editing
- pegRNA
Prime editing guide RNA
- PEI
Polyethyleneimine
- PK
Pharmacokinetic
- PKR
RNA-dependent protein kinase
- Plk1
Polo-like kinase-1
- PLNP
Phospholipid-modified cationic lipid nanoparticle
- RRs
Pattern recognition receptors
- PS
Phosphonothioate
- rAAV
Recombinant AAV
- rAAV2-CFTR
Recombinant AAV2 capsid RBCs Red blood cells
- RCR
Replication-competent retroviral vectors
- Rig-I
Retinoic acid-inducible gene I
- RNAi
RNA interference
- RNP
Ribonucleoprotein
- RT
Reverse transcriptase
- SaCas9
Staphylococcus aureus Cas9
- SARS-CoV-2
Severe acute respiratory syndrome coronavirus 2
- SauriCas9
Staphylococcus auricularis Cas9
- SC
Subcutaneous
- SEC
Size exclusion chromatography
- sgRNA
Single-guide RNA
- siRNA
Small interfering RNA
- SLNPs
Solid LNPs
- SMRTs
Specific modified mRNA translation systems
- SNAs
Spherical nucleic acids
- SORT
Selective organ targeting
- SRSRs
Short regularly- spaced repeats
- ssODN
Single-stranded DNA oligonucleotides
- STR-R8
Stearylated octa-arginine
- SUVs
Small unilamellar vesicles
- T2D
Type 2 diabetes
- TALENs
Transcription activator-like effector nucleases
- TCR
T-cell receptors
- TEM
Transmission electron microscopy
- TFR
Total flow rate
- TLRs
Toll-like receptors
- Tm
Transition temperature
- Tmc1
Transmembrane channel-like 1
- trcrRNA
Trans-activating CRISPR RNA
- UCART19
Universal Chimeric Antigen Receptor 19 T-cells
- UNAs
Unlocked nucleic acids
- VLPs
Virus-like Particles
- VNB
Vapor nanobubble
- ZFNs
Zinc-finger nucleases
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest statement. Dr. Howard Gendelman is a co-founder of Exavir Therapeutics. Inc. The opinions expressed are his own.
Declaration of Competing Interest
The authors have no known competing financial interests or personal relationships that would have influence what is reported in this paper.
References
- 1.Capecchi MR. Altering the genome by homologous recombination. Science 1989; 244(4910): 1288–92. [DOI] [PubMed] [Google Scholar]
- 2.Mario R Capecchi SMJE, Oliver Smithies. The Nobel Prize in Physiology or Medicine 2007. 2007. The Nobel Prize in Physiology or Medicine 2007 (accessed JUNE 20 2022).
- 3.Rouet P, Smih F, Jasin M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Molecular and cellular biology 1994; 14(12): 8096–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 2012; 337(6096): 816–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cullis PR, Hope MJ. Lipid Nanoparticle Systems for Enabling Gene Therapies. Molecular Therapy 2017; 25(7): 1467–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kim Y-G, Cha J, Chandrasegaran S. Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proceedings of the National Academy of Sciences USA 1996; 93(3): 1156–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chandrasegaran S Recent advances in the use of ZFN-mediated gene editing for human gene therapy. Cell & gene therapy insights 2017; 3(1): 33–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Carroll D, Morton JJ, Beumer KJ, Segal DJ. Design, construction and in vitro testing of zinc finger nucleases. Nature Protocols 2006; 1(3): 1329–41. [DOI] [PubMed] [Google Scholar]
- 9.Qiu M, Glass Z, Chen J, et al. Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves liver-specific in vivo genome editing of Angptl3. Proceedings of the National Academy of Sciences 2021; 118(10): e2020401118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Miller JC, Tan S, Qiao G, et al. A TALE nuclease architecture for efficient genome editing. Nature Biotechnology 2011; 29(2): 143–8. [DOI] [PubMed] [Google Scholar]
- 11.Paschon DE, Lussier S, Wangzor T, et al. Diversifying the structure of zinc finger nucleases for high-precision genome editing. Nature Communications 2019; 10(1): 1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Perez EE, Wang J, Miller JC, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nature biotechnology 2008; 26(7): 808–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Becker S, Boch J. TALE and TALEN genome editing technologies. Gene and Genome Editing 2021; 2: 100007. [Google Scholar]
- 14.Kubik G, Summerer D. Achieving single-nucleotide resolution of 5-methylcytosine detection with TALEs. Chembiochem 2015; 16(2): 228–31. [DOI] [PubMed] [Google Scholar]
- 15.Boissel S, Jarjour J, Astrakhan A, et al. megaTALs: a rare-cleaving nuclease architecture for therapeutic genome engineering. Nucleic acids research 2014; 42(4): 2591–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kleinstiver BP, Wang L, Wolfs JM, et al. The I-TevI nuclease and linker domains contribute to the specificity of monomeric TALENs. G3 (Bethesda) 2014; 4(6): 1155–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kim Y, Kweon J, Kim J-S. TALENs and ZFNs are associated with different mutation signatures. Nature Methods 2013; 10(3): 185-. [DOI] [PubMed] [Google Scholar]
- 18.Qasim W, Zhan H, Samarasinghe S, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Science Translational Medicine 2017; 9(374). [DOI] [PubMed] [Google Scholar]
- 19.Mullally G, Van Aelst K, Naqvi MM, et al. 5′ modifications to CRISPR–Cas9 gRNA can change the dynamics and size of R-loops and inhibit DNA cleavage. Nucleic acids research 2020; 48(12): 6811–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cox DBT, Platt RJ, Zhang F. Therapeutic genome editing: prospects and challenges. Nature Medicine 2015; 21(2): 121–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cui Z, Liu H, Zhang H, et al. The comparison of ZFNs, TALENs, and SpCas9 by GUIDE-seq in HPV-targeted gene therapy. Molecular therapy Nucleic acids 2021; 26: 1466–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Kaur H, Bruno JG, Kumar A, Sharma TK. Aptamers in the Therapeutics and Diagnostics Pipelines. Theranostics 2018; 8(15): 4016–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ishino Y, Shinagawa H, Makino K, Amemura M, Nakata A. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. Journal of Bacteriology 1987; 169(12): 5429–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Groenen PM, Bunschoten AE, van Soolingen D, van Embden JD. Nature of DNA polymorphism in the direct repeat cluster of Mycobacterium tuberculosis; application for strain differentiation by a novel typing method. Molecular Microbiology 1993; 10(5): 1057–65. [DOI] [PubMed] [Google Scholar]
- 25.Mojica FJM, Díez-Villaseñor C, Soria E, Juez G. Biological significance of a family of regularly spaced repeats in the genomes of Archaea, Bacteria and mitochondria. Molecular Microbiology 2000; 36(1): 244–6. [DOI] [PubMed] [Google Scholar]
- 26.Jansen R, Embden JD, Gaastra W, Schouls LM. Identification of genes that are associated with DNA repeats in prokaryotes. Molecular Microbiology 2002; 43(6): 1565–75. [DOI] [PubMed] [Google Scholar]
- 27.Mojica FJM, Rodriguez-Valera F. The discovery of CRISPR in archaea and bacteria. The FEBS Journal 2016; 283(17): 3162–9. [DOI] [PubMed] [Google Scholar]
- 28.Haft DH, Selengut J, Mongodin EF, Nelson KE. A guild of 45 CRISPR-associated (Cas) protein families and multiple CRISPR/Cas subtypes exist in prokaryotic genomes. PLoS computational biology 2005; 1(6): e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Bolotin A, Quinquis B, Sorokin A, Ehrlich SD. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology (Reading) 2005; 151(Pt 8): 2551–61. [DOI] [PubMed] [Google Scholar]
- 30.Mojica FJ, Díez-Villaseñor C, García-Martínez J, Soria E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. Journal of Molecular Evolultion 2005; 60(2): 174–82. [DOI] [PubMed] [Google Scholar]
- 31.Makarova KS, Grishin NV, Shabalina SA, Wolf YI, Koonin EV. A putative RNA-interference-based immune system in prokaryotes: computational analysis of the predicted enzymatic machinery, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biology Direct 2006; 1(1): 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Barrangou R, Fremaux C, Deveau H, et al. CRISPR Provides Acquired Resistance Against Viruses in Prokaryotes. Science 2007; 315(5819): 1709–12. [DOI] [PubMed] [Google Scholar]
- 33.Brouns SJ, Jore MM, Lundgren M, et al. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 2008; 321(5891): 960–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Deltcheva E, Chylinski K, Sharma CM, et al. CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 2011; 471(7340): 602–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gasiunas G, Barrangou R, Horvath P, Siksnys V. Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proceedings of the National Academy of Sciences USA 2012; 109(39): E2579–E86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013; 339(6121): 819–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Hwang WY, Fu Y, Reyon D, et al. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nature biotechnology 2013; 31(3): 227–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Crystal RG. Adenovirus: the first effective in vivo gene delivery vector. Human gene therapy 2014; 25(1): 3–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tang X, Lowder LG, Zhang T, et al. A CRISPR-Cpf1 system for efficient genome editing and transcriptional repression in plants. Nat Plants 2017; 3: 17018. [DOI] [PubMed] [Google Scholar]
- 40.Makarova KS, Wolf YI, Koonin EV. Classification and Nomenclature of CRISPR-Cas Systems: Where from Here? The CRISPR journal 2018; 1(5): 325–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Adli M The CRISPR tool kit for genome editing and beyond. Nature Communications 2018; 9(1): 1911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pfeiffer P, Goedecke W, Obe G. Mechanisms of DNA double-strand break repair and their potential to induce chromosomal aberrations. Mutagenesis 2000; 15(4): 289–302. [DOI] [PubMed] [Google Scholar]
- 43.Gallagher DN, Haber JE. Repair of a Site-Specific DNA Cleavage: Old-School Lessons for Cas9-Mediated Gene Editing. ACS Chemical Biology 2018; 13(2): 397–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Yip BH. Recent Advances in CRISPR/Cas9 Delivery Strategies. Biomolecules 2020; 10(6): 839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Luo YL, Xu CF, Li HJ, et al. Macrophage-Specific in Vivo Gene Editing Using Cationic Lipid-Assisted Polymeric Nanoparticles. ACS Nano 2018; 12(2): 994–1005. [DOI] [PubMed] [Google Scholar]
- 46.Di Stazio M, Foschi N, Athanasakis E, Gasparini P, d’Adamo AP. Systematic analysis of factors that improve homologous direct repair (HDR) efficiency in CRISPR/Cas9 technique. Plos one 2021; 16(3): e0247603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wasels F, Jean-Marie J, Collas F, López-Contreras AM, Lopes Ferreira N. A two-plasmid inducible CRISPR/Cas9 genome editing tool for Clostridium acetobutylicum. Journal of Microbiological Methods 2017; 140: 5–11. [DOI] [PubMed] [Google Scholar]
- 48.Wu Y, Zhou H, Fan X, et al. Correction of a genetic disease by CRISPR-Cas9-mediated gene editing in mouse spermatogonial stem cells. Cell Research 2015; 25(1): 67–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Deng S, Li X, Liu S, et al. Codelivery of CRISPR-Cas9 and chlorin e6 for spatially controlled tumor-specific gene editing with synergistic drug effects. Science Advances 2020; 6(29): eabb4005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Albers J, Danzer C, Rechsteiner M, et al. A versatile modular vector system for rapid combinatorial mammalian genetics. Journal of Clinical Investigation 2015; 125(4): 1603–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kabadi AM, Ousterout DG, Hilton IB, Gersbach CA. Multiplex CRISPR/Cas9-based genome engineering from a single lentiviral vector. Nucleic Acids Res 2014; 42(19): e147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kamimura K, Yokoo T, Abe H, et al. Image-Guided Hydrodynamic Gene Delivery: Current Status and Future Directions. Pharmaceutics 2015; 7(3): 213–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wilbie D, Walther J, Mastrobattista E. Delivery Aspects of CRISPR/Cas for in Vivo Genome Editing. Acc Chem Res 2019; 52(6): 1555–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lino CA, Harper JC, Carney JP, Timlin JA. Delivering CRISPR: a review of the challenges and approaches. Drug Delivery 2018; 25(1): 1234–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sun M, Wang T, Li L, et al. The Application of Inorganic Nanoparticles in Molecular Targeted Cancer Therapy: EGFR Targeting. Frontiers in pharmacology 2021; 12: 702445-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zuris JA, Thompson DB, Shu Y, et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nature Biotechnology 2015; 33(1): 73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chen S, Lee B, Lee AY, Modzelewski AJ, He L. Highly Efficient Mouse Genome Editing by CRISPR Ribonucleoprotein Electroporation of Zygotes. J Biol Chem 2016; 291(28): 14457–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Raes L, Pille M, Harizaj A, et al. Cas9 RNP transfection by vapor nanobubble photoporation for ex vivo cell engineering. Molecular Therapy-Nucleic Acids 2021; 25: 696–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vaidyanathan S, Azizian KT, Haque A, et al. Uridine Depletion and Chemical Modification Increase Cas9 mRNA Activity and Reduce Immunogenicity without HPLC Purification. Molecular Therapy Nucleic Acids 2018; 12: 530–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Karikó K, Muramatsu H, Keller JM, Weissman D. Increased erythropoiesis in mice injected with submicrogram quantities of pseudouridine-containing mRNA encoding erythropoietin. Molecular Therapy 2012; 20(5): 948–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kelley ML, Strezoska Ž, He K, Vermeulen A, Smith A. Versatility of chemically synthesized guide RNAs for CRISPR-Cas9 genome editing. Journal of Biotechnology 2016; 233: 74–83. [DOI] [PubMed] [Google Scholar]
- 62.Mu W, Zhang Y, Xue X, Liu L, Wei X, Wang H. 5’ capped and 3’ polyA-tailed sgRNAs enhance the efficiency of CRISPR-Cas9 system. Protein Cell 2019; 10(3): 223–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kocak DD, Josephs EA, Bhandarkar V, Adkar SS, Kwon JB, Gersbach CA. Increasing the specificity of CRISPR systems with engineered RNA secondary structures. Nature Biotechnol 2019; 37(6): 657–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Basila M, Kelley ML, Smith AVB. Minimal 2’-O-methyl phosphorothioate linkage modification pattern of synthetic guide RNAs for increased stability and efficient CRISPR-Cas9 gene editing avoiding cellular toxicity. PLoS One 2017; 12(11): e0188593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lee CS, Bishop ES, Zhang R, et al. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes & diseases 2017; 4(2): 43–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lapinaite A, Doudna JA, Cate JH. Programmable RNA recognition using a CRISPR-associated Argonaute. Proceedings of the National Academy of Sciences USA 2018; 115(13): 3368–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Schubert MS, Cedrone E, Neun B, Behlke MA, Dobrovolskaia MA. Chemical Modification of CRISPR gRNAs Eliminate type I Interferon Responses in Human Peripheral Blood Mononuclear Cells. J Cytokine Biol 2018; 3(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nahar S, Sehgal P, Azhar M, et al. A G-quadruplex motif at the 3′ end of sgRNAs improves CRISPR–Cas9 based genome editing efficiency. Chemical Communications 2018; 54(19): 2377–80. [DOI] [PubMed] [Google Scholar]
- 69.Moon SB, Kim DY, Ko JH, Kim JS, Kim YS. Improving CRISPR Genome Editing by Engineering Guide RNAs. Trends Biotechnol 2019; 37(8): 870–81. [DOI] [PubMed] [Google Scholar]
- 70.Vidigal JA, Ventura A. Rapid and efficient one-step generation of paired gRNA CRISPR-Cas9 libraries. Nature Communications 2015; 6: 8083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Taemaitree L, Shivalingam A, El-Sagheer AH, Brown T. An artificial triazole backbone linkage provides a split-and-click strategy to bioactive chemically modified CRISPR sgRNA. Nature Communications 2019; 10(1): 1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mir A, Alterman JF, Hassler MR, et al. Heavily and fully modified RNAs guide efficient SpyCas9-mediated genome editing. Nature Communications 2018; 9(1): 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hendel A, Bak RO, Clark JT, et al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nature Biotechnology 2015; 33(9): 985–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Scott T, Soemardy C, Morris KV. Development of a facile approach for generating chemically modified CRISPR/Cas9 RNA. Molecular Therapy-Nucleic Acids 2020; 19: 1176–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Allen D, Rosenberg M, Hendel A. Using Synthetically Engineered Guide RNAs to Enhance CRISPR Genome Editing Systems in Mammalian Cells. Frontiers in Genome Editing 2021; 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Phua KK, Leong KW, Nair SK. Transfection efficiency and transgene expression kinetics of mRNA delivered in naked and nanoparticle format. Journal of Controlled Release 2013; 166(3): 227–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Miller JB, Zhang S, Kos P, et al. Non-Viral CRISPR/Cas Gene Editing In Vitro and In Vivo Enabled by Synthetic Nanoparticle Co-Delivery of Cas9 mRNA and sgRNA. Angew Chem Int Ed Engl 2017; 56(4): 1059–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Liu G, Lin Q, Jin S, Gao C. The CRISPR-Cas toolbox and gene editing technologies. Molecular Cell 2021. [DOI] [PubMed]
- 79.Wu S-S, Li Q-C, Yin C-Q, Xue W, Song C-Q. Advances in CRISPR/Cas-based gene therapy in human genetic diseases. Theranostics 2020; 10(10): 4374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Pruett-Miller SM. Assessing off-target editing of CRISPR-cas9 systems. The CRISPR Journal 2020; 3(6): 430–2. [DOI] [PubMed] [Google Scholar]
- 81.Tsai SQ, Zheng Z, Nguyen NT, et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nature biotechnology 2015; 33(2): 187–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tsai SQ, Nguyen NT, Malagon-Lopez J, Topkar VV, Aryee MJ, Joung JK. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR–Cas9 nuclease off-targets. Nature Methods 2017; 14(6): 607–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cebrian-Serrano A, Davies B. CRISPR-Cas orthologues and variants: optimizing the repertoire, specificity and delivery of genome engineering tools. Mammalian Genome 2017; 28(7): 247–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kampmann M CRISPRi and CRISPRa Screens in Mammalian Cells for Precision Biology and Medicine. ACS chemical biology 2018; 13(2): 406–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Gilbert LA, Larson MH, Morsut L, et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013; 154(2): 442–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gaudelli NM, Komor AC, Rees HA, et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017; 551(7681): 464–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Komor AC, Kim YB, Packer MS, Zuris JA, Liu DR. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016; 533(7603): 420–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Scholefield J, Harrison PT. Prime editing – an update on the field. Gene Therapy 2021; 28(7): 396–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Anzalone AV, Randolph PB, Davis JR, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019; 576(7785): 149–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Heckl D, Kowalczyk MS, Yudovich D, et al. Generation of mouse models of myeloid malignancy with combinatorial genetic lesions using CRISPR-Cas9 genome editing. Nat Biotechnol 2014; 32(9): 941–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Friedmann T The future for gene therapy--a reevaluation. Ann N Y Acad Sci 1976; 265: 141–52. [DOI] [PubMed] [Google Scholar]
- 92.Rittiner JE, Moncalvo M, Chiba-Falek O, Kantor B. Gene-Editing Technologies Paired With Viral Vectors for Translational Research Into Neurodegenerative Diseases. Frontiers in Molecular Neuroscience 2020; 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Bulcha JT, Wang Y, Ma H, Tai PWL, Gao G. Viral vector platforms within the gene therapy landscape. Signal Transduction and Targeted Therapy 2021; 6(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Emily Capra AG, Alberto Loche, and Jeff Smith. Gene-therapy innovation: Unlocking the promise of viral vectors 2021. https://www.mckinsey.com/industries/life-sciences/our-insights/gene-therapy-innovation-unlocking-the-promise-of-viral-vectors (accessed June 10 2022).
- 95.Almarza D, Bussadori G, Navarro M, Mavilio F, Larcher F, Murillas R. Risk assessment in skin gene therapy: viral–cellular fusion transcripts generated by proviral transcriptional read-through in keratinocytes transduced with self-inactivating lentiviral vectors. Gene Therapy 2011; 18(7): 674–81. [DOI] [PubMed] [Google Scholar]
- 96.Ertl HCJ. Immunogenicity and toxicity of AAV gene therapy. Frontiers of Immunology 2022; 13: 975803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hanawa H, Kelly PF, Nathwani AC, et al. Comparison of various envelope proteins for their ability to pseudotype lentiviral vectors and transduce primitive hematopoietic cells from human blood. Molecular Therapy 2002; 5(3): 242–51. [DOI] [PubMed] [Google Scholar]
- 98.Bieniasz PD, Grdina TA, Bogerd HP, Cullen BR. Recruitment of cyclin T1/P-TEFβ to an HIV type 1 long terminal repeat promoter proximal RNA target is both necessary and sufficient for full activation of transcription. Proceeding of the National Academy of Sciences USA 1999; 96(14): 7791–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Neville M, Stutz F, Lee L, Davis LI, Rosbash M. The importin-beta family member Crm1p bridges the interaction between Rev and the nuclear pore complex during nuclear export. Current Biology 1997; 7(10): 767–75. [DOI] [PubMed] [Google Scholar]
- 100.Seissler T, Marquet R, Paillart J-C. Hijacking of the Ubiquitin/Proteasome Pathway by the HIV Auxiliary Proteins. Viruses 2017; 9(11): 322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bear AS, Morgan RA, Cornetta K, et al. Replication-competent retroviruses in gene-modified T cells used in clinical trials: is it time to revise the testing requirements? Molecular Therapy 2012; 20(2): 246–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Milone MC, O’Doherty U. Clinical use of lentiviral vectors. Leukemia 2018; 32(7): 1529–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Sabnis S, Kumarasinghe ES, Salerno T, et al. A Novel Amino Lipid Series for mRNA Delivery: Improved Endosomal Escape and Sustained Pharmacology and Safety in Non-human Primates. Mol Ther 2018; 26(6): 1509–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kim HS, Hwang G-H, Lee HK, et al. CReVIS-Seq: A highly accurate and multiplexable method for genome-wide mapping of lentiviral integration sites. Molecular Therapy-Methods & Clinical Development 2021; 20: 792–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tian J, Andreadis ST. Independent and high-level dual-gene expression in adult stem-progenitor cells from a single lentiviral vector. Gene Ther 2009; 16(7): 874–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Gallardo J, Pérez-Illana M, Martín-González N, San Martín C. Adenovirus Structure: What Is New? International journal of molecular sciences 2021; 22(10): 5240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yang ZR, Wang HF, Zhao J, et al. Recent developments in the use of adenoviruses and immunotoxins in cancer gene therapy. Cancer Gene Therapy 2007; 14(7): 599–615. [DOI] [PubMed] [Google Scholar]
- 108.Pérez-Berná AJ, Marion S, Chichón FJ, et al. Distribution of DNA-condensing protein complexes in the adenovirus core. Nucleic acids research 2015; 43(8): 4274–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Alba R, Bosch A, Chillon M. Gutless adenovirus: last-generation adenovirus for gene therapy. Gene Therapy 2005; 12(1): S18–S27. [DOI] [PubMed] [Google Scholar]
- 110.Rosengart TK, Lee LY, Patel SR, et al. Angiogenesis gene therapy: phase I assessment of direct intramyocardial administration of an adenovirus vector expressing VEGF121 cDNA to individuals with clinically significant severe coronary artery disease. Circulation 1999; 100(5): 468–74. [DOI] [PubMed] [Google Scholar]
- 111.Raper SE, Yudkoff M, Chirmule N, et al. A pilot study of in vivo liver-directed gene transfer with an adenoviral vector in partial ornithine transcarbamylase deficiency. Hum Gene Ther 2002; 13(1): 163–75. [DOI] [PubMed] [Google Scholar]
- 112.Wang D, Mou H, Li S, et al. Adenovirus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses. Human gene therapy 2015; 26(7): 432–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lin J, Wong K-C. Off-target predictions in CRISPR-Cas9 gene editing using deep learning. Bioinformatics 2018; 34(17): i656–i63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Bjursell M, Porritt MJ, Ericson E, et al. Therapeutic Genome Editing With CRISPR/Cas9 in a Humanized Mouse Model Ameliorates α1-antitrypsin Deficiency Phenotype. EBioMedicine 2018; 29: 104–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Wolins N, Lozier J, Eggerman T, Jones E, Aguilar‐Cordova E, Vostal J. Intravenous administration of replication‐incompetent adenovirus to rhesus monkeys induces thrombocytopenia by increasing in vivo platelet clearance. British journal of haematology 2003; 123(5): 903–5. [DOI] [PubMed] [Google Scholar]
- 116.Koo T, Yoon A-R, Cho H-Y, Bae S, Yun C-O, Kim J-S. Selective disruption of an oncogenic mutant allele by CRISPR/Cas9 induces efficient tumor regression. Nucleic Acids Research 2017; 45(13): 7897–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Nwanegbo E, Vardas E, Gao W, et al. Prevalence of neutralizing antibodies to adenoviral serotypes 5 and 35 in the adult populations of The Gambia, South Africa, and the United States. Clinical and diagnostic laboratory immunology 2004; 11(2): 351–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Atchison RW, Casto BC, Hammon WM. ADENOVIRUS-ASSOCIATED DEFECTIVE VIRUS PARTICLES. Science 1965; 149(3685): 754–6. [DOI] [PubMed] [Google Scholar]
- 119.Lusby E, Fife KH, Berns KI. Nucleotide sequence of the inverted terminal repetition in adeno-associated virus DNA. Journal of virology 1980; 34(2): 402–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Swingle KL, Billingsley MM, Bose SK, et al. Amniotic fluid stabilized lipid nanoparticles for in utero intra-amniotic mRNA delivery. Journal of Controlled Release 2022; 341: 616–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gao G, Vandenberghe LH, Alvira MR, et al. Clades of Adeno-Associated Viruses Are Widely Disseminated in Human Tissues. Journal of Virology 2004; 78(12): 6381–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Flotte T, Carter B, Conrad C, et al. A Phase I Study of an Adeno-Associated Virus-CFTR Gene Vector in Adult CF Patients with Mild Lung Disease. Johns Hopkins Children’s Center, Baltimore, Maryland. Human Gene Therapy 1996; 7(9): 1145–59. [DOI] [PubMed] [Google Scholar]
- 123.Knipe D, Howley P, Griffin D, et al. Fields Virology, Volumes 1 and 2 2013.
- 124.Duan D, Yue Y, Yan Z, Engelhardt JF. A new dual-vector approach to enhance recombinant adeno-associated virus-mediated gene expression through intermolecular cis activation. Nature Medicine 2000; 6(5): 595–8. [DOI] [PubMed] [Google Scholar]
- 125.Davis KM, Pattanayak V, Thompson DB, Zuris JA, Liu DR. Small molecule–triggered Cas9 protein with improved genome-editing specificity. Nature Chemical Biology 2015; 11(5): 316–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dow LE, Fisher J, O’Rourke KP, et al. Inducible in vivo genome editing with CRISPR-Cas9. Nature Biotechnology 2015; 33(4): 390–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Colella P, Ronzitti G, Mingozzi F. Emerging issues in AAV-mediated in vivo gene therapy. Molecular Therapy-Methods & Clinical Development 2018; 8: 87–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Levy JM, Yeh W-H, Pendse N, et al. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nature Biomedical Engineering 2020; 4(1): 97–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Maurya S, Sarangi P, Jayandharan GR. Safety of Adeno-associated virus-based vector-mediated gene therapy—impact of vector dose. Cancer Gene Therapy 2022. [DOI] [PubMed]
- 130.Shahryari A, Saghaeian Jazi M, Mohammadi S, et al. Development and Clinical Translation of Approved Gene Therapy Products for Genetic Disorders. Frontiers in Genetics 2019; 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Noad R, Roy P. Virus-like particles as immunogens. Trends in Microbiology 2003; 11(9): 438–44. [DOI] [PubMed] [Google Scholar]
- 132.Banskota S, Raguram A, Suh S, et al. Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell 2022; 185(2): 250–65.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Choi JG, Dang Y, Abraham S, et al. Lentivirus pre-packed with Cas9 protein for safer gene editing. Gene Ther 2016; 23(7): 627–33. [DOI] [PubMed] [Google Scholar]
- 134.Mangeot PE, Risson V, Fusil F, et al. Genome editing in primary cells and in vivo using viral-derived Nanoblades loaded with Cas9-sgRNA ribonucleoproteins. Nature Communications 2019; 10(1): 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lan T, Que H, Luo M, Zhao X, Wei X. Genome editing via non-viral delivery platforms: current progress in personalized cancer therapy. Mol Cancer 2022; 21(1): 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ramamoorth M, Narvekar A. Non viral vectors in gene therapyan overview. J Clin Diagn Res 2015; 9(1): GE01–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Zu H, Gao D. Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS J 2021; 23(4): 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Verbeke R, Lentacker I, De Smedt SC, Dewitte H. Three decades of messenger RNA vaccine development. Nano Today 2019; 28: 100766. [Google Scholar]
- 139.2- Modification of drug release. In: Bruschi ML, ed. Strategies to Modify the Drug Release from Pharmaceutical Systems: Woodhead Publishing; 2015: 15–28. [Google Scholar]
- 140.Begines B, Ortiz T, Pérez-Aranda M, et al. Polymeric Nanoparticles for Drug Delivery: Recent Developments and Future Prospects. Nanomaterials (Basel, Switzerland) 2020; 10(7): 1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Cano A, Sánchez-López E, Ettcheto M, et al. Current advances in the development of novel polymeric nanoparticles for the treatment of neurodegenerative diseases. Nanomedicine 2020; 15(12): 1239–61. [DOI] [PubMed] [Google Scholar]
- 142.Masri A, Anwar A, Khan NA, Siddiqui R. The Use of Nanomedicine for Targeted Therapy against Bacterial Infections. Antibiotics 2019; 8(4): 260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Parhi P, Mohanty C, Sahoo SK. Nanotechnology-based combinational drug delivery: an emerging approach for cancer therapy. Drug Discovery Today 2012; 17(17): 1044–52. [DOI] [PubMed] [Google Scholar]
- 144.Longmire MR, Ogawa M, Choyke PL, Kobayashi H. Biologically optimized nanosized molecules and particles: more than just size. Bioconjugate chemistry 2011; 22(6): 993–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Longmire M, Choyke PL, Kobayashi H. Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats 2008. [DOI] [PMC free article] [PubMed]
- 146.De Jong WH, Borm PJA. Drug delivery and nanoparticles:applications and hazards. International journal of nanomedicine 2008; 3(2): 133–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wolff JA, Rozema DB. Breaking the bonds: non-viral vectors become chemically dynamic. Molecular Therapy 2008; 16(1): 8–15. [DOI] [PubMed] [Google Scholar]
- 148.Tros de Ilarduya C, Sun Y, Düzgüneş N. Gene delivery by lipoplexes and polyplexes. Eur J Pharm Sci 2010; 40(3): 159–70. [DOI] [PubMed] [Google Scholar]
- 149.Rozema DB, Lewis DL, Wakefield DH, et al. Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci U S A 2007; 104(32): 12982–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lorenzer C, Dirin M, Winkler A-M, Baumann V, Winkler J. Going beyond the liver: Progress and challenges of targeted delivery of siRNA therapeutics. Journal of Controlled Release 2015; 203: 1–15. [DOI] [PubMed] [Google Scholar]
- 151.Roberts TC, Langer R, Wood MJA. Advances in oligonucleotide drug delivery. Nature Reviews Drug Discovery 2020; 19(10): 673–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Brader ML, Williams SJ, Banks JM, Hui WH, Zhou ZH, Jin L. Encapsulation state of messenger RNA inside lipid nanoparticles. Biophysical Journal 2021; 120(14): 2766–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kon E, Elia U, Peer D. Principles for designing an optimal mRNA lipid nanoparticle vaccine. Current Opinion in Biotechnology 2022; 73: 329–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Eygeris Y, Patel S, Jozic A, Sahay G. Deconvoluting lipid nanoparticle structure for messenger RNA delivery. Nano letters 2020; 20(6): 4543–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Chapter Kumar R. 8 - Lipid-Based Nanoparticles for Drug-Delivery Systems. In: Mohapatra SS, Ranjan S, Dasgupta N, Mishra RK, Thomas S, eds. Nanocarriers for Drug Delivery: Elsevier; 2019: 249–84. [Google Scholar]
- 156.Rajabi M A Mousa S Lipid Nanoparticles and their Application in Nanomedicine. Current Pharmaceutical Biotechnology 2016; 17(8): 662–72. [DOI] [PubMed] [Google Scholar]
- 157.Zhao Y, Huang L. Chapter Two - Lipid Nanoparticles for Gene Delivery. In: Huang L, Liu D, Wagner E, eds. Advances in Genetics: Academic Press; 2014: 13–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hafez I, Maurer N, Cullis P. On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids. Gene therapy 2001; 8(15): 1188–96. [DOI] [PubMed] [Google Scholar]
- 159.Zhi D, Zhang S, Cui S, Zhao Y, Wang Y, Zhao D. The headgroup evolution of cationic lipids for gene delivery. Bioconjugate chemistry 2013; 24(4): 487–519. [DOI] [PubMed] [Google Scholar]
- 160.Semple SC, Akinc A, Chen J, et al. Rational design of cationic lipids for siRNA delivery. Nature biotechnology 2010; 28(2): 172–6. [DOI] [PubMed] [Google Scholar]
- 161.Alabi CA, Love KT, Sahay G, et al. Multiparametric approach for the evaluation of lipid nanoparticles for siRNA delivery. Proceedings of the National Academy of Sciences 2013; 110(32): 12881–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Tam YYC, Chen S, Cullis PR. Advances in lipid nanoparticles for siRNA delivery. Pharmaceutics 2013; 5(3): 498–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Finn JD, Smith AR, Patel MC, et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell reports 2018; 22(9): 2227–35. [DOI] [PubMed] [Google Scholar]
- 164.Malone RW, Felgner PL, Verma IM. Cationic liposome-mediated RNA transfection. Proceedings of the National Academy of Sciences 1989; 86(16): 6077–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Song S, Shen Z, Chen L, Brayman AA, Miao CH. Explorations of high-intensity therapeutic ultrasound and microbubble-mediated gene delivery in mouse liver. Gene Ther 2011; 18(10): 1006–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Fehring V, Schaeper U, Ahrens K, et al. Delivery of therapeutic siRNA to the lung endothelium via novel lipoplex formulation DACC. Molecular Therapy 2014; 22(4): 811–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Ross PC, Hui SW. Lipoplex size is a major determinant of in vitro lipofection efficiency. Gene Therapy 1999; 6(4): 651–9. [DOI] [PubMed] [Google Scholar]
- 168.Filion MC, Phillips NC. Toxicity and immunomodulatory activity of liposomal vectors formulated with cationic lipids toward immune effector cells. Biochim Biophys Acta 1997; 1329(2): 345–56. [DOI] [PubMed] [Google Scholar]
- 169.Peer D Immunotoxicity derived from manipulating leukocytes with lipid-based nanoparticles. Advanced drug delivery reviews 2012; 64(15): 1738–48. [DOI] [PubMed] [Google Scholar]
- 170.Heyes J, Palmer L, Bremner K, MacLachlan I. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. Journal of controlled release 2005; 107(2): 276–87. [DOI] [PubMed] [Google Scholar]
- 171.Hassett KJ, Benenato KE, Jacquinet E, et al. Optimization of Lipid Nanoparticles for Intramuscular Administration of mRNA Vaccines. Molecular Therapy - Nucleic Acids 2019; 15: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Whitehead KA, Sahay G, Li GZ, et al. Synergistic Silencing: Combinations of Lipid-like Materials for Efficacious siRNA Delivery. Molecular Therapy 2011; 19(9): 1688–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Felgner PL, Gadek TR, Holm M, et al. Lipofection: a highly efficient, lipid-mediated DNA-transfection procedure. Proceedings of the National Academy of Sciences 1987; 84(21): 7413–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Hattori Y, Suzuki S, Kawakami S, Yamashita F, Hashida M. The role of dioleoylphosphatidylethanolamine (DOPE) in targeted gene delivery with mannosylated cationic liposomes via intravenous route. Journal of Controlled Release 2005; 108(2): 484–95. [DOI] [PubMed] [Google Scholar]
- 175.Barenholz Y Doxil® — The first FDA-approved nano-drug: Lessons learned. Journal of Controlled Release 2012; 160(2): 117–34. [DOI] [PubMed] [Google Scholar]
- 176.Wang F, Li C, Cheng J, Yuan Z. Recent advances on inorganic nanoparticle-based cancer therapeutic agents. International journal of environmental research and public health 2016; 13(12): 1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Leung AK, Tam YYC, Chen S, Hafez IM, Cullis PR. Microfluidic mixing: a general method for encapsulating macromolecules in lipid nanoparticle systems. The Journal of Physical Chemistry B 2015; 119(28): 8698–706. [DOI] [PubMed] [Google Scholar]
- 178.Kauffman KJ, Dorkin JR, Yang JH, et al. Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with fractional factorial and definitive screening designs. Nano letters 2015; 15(11): 7300–6. [DOI] [PubMed] [Google Scholar]
- 179.Bernsdorff C, Winter R. Differential properties of the sterols cholesterol, ergosterol, β-sitosterol, trans-7-dehydrocholesterol, stigmasterol and lanosterol on DPPC bilayer order. The Journal of Physical Chemistry B 2003; 107(38): 10658–64. [Google Scholar]
- 180.Takahashi H, Sinoda K, Hatta I. Effects of cholesterol on the lamellar and the inverted hexagonal phases of dielaidoylphosphatidylethanolamine. Biochimica et Biophysica Acta (BBA) - General Subjects 1996; 1289(2): 209–16. [DOI] [PubMed] [Google Scholar]
- 181.Sakurai F, Nishioka T, Yamashita F, Takakura Y, Hashida M. Effects of erythrocytes and serum proteins on lung accumulation of lipoplexes containing cholesterol or DOPE as a helper lipid in the single-pass rat lung perfusion system. European Journal of Pharmaceutics and Biopharmaceutics 2001; 52(2): 165–72. [DOI] [PubMed] [Google Scholar]
- 182.Pozzi D, Marchini C, Cardarelli F, et al. Transfection efficiency boost of cholesterol-containing lipoplexes. Biochimica et Biophysica Acta (BBA) - Biomembranes 2012; 1818(9): 2335–43. [DOI] [PubMed] [Google Scholar]
- 183.Pilkington EH, Suys EJA, Trevaskis NL, et al. From influenza to COVID-19: Lipid nanoparticle mRNA vaccines at the frontiers of infectious diseases. Acta Biomaterialia 2021; 131: 16–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Torchilin VP, Trubetskoy VS. Which polymers can make nanoparticulate drug carriers long-circulating? Advanced Drug Delivery Reviews 1995; 16(2): 141–55. [Google Scholar]
- 185.Allen TM. The use of glycolipids and hydrophilic polymers in avoiding rapid uptake of liposomes by the mononuclear phagocyte system. Advanced Drug Delivery Reviews 1994; 13(3): 285–309. [Google Scholar]
- 186.Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Advanced Drug Delivery Reviews 2016; 99: 28–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Schlich M, Palomba R, Costabile G, et al. Cytosolic delivery of nucleic acids: The case of ionizable lipid nanoparticles. Bioengineering & Translational Medicine 2021; 6(2): e10213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.MacLachlan I Liposomal formulations for nucleic acid delivery. Antisense Drug Technology: CRC press; 2007: 255–88.
- 189.Jahn A, Vreeland WN, Gaitan M, Locascio LE. Controlled Vesicle Self-Assembly in Microfluidic Channels with Hydrodynamic Focusing. Journal of the American Chemical Society 2004; 126(9): 2674–5. [DOI] [PubMed] [Google Scholar]
- 190.Wagner A, Vorauer-Uhl K, Kreismayr G, Katinger H. The crossflow injection technique: an improvement of the ethanol injection method. J Liposome Res 2002; 12(3): 259–70. [DOI] [PubMed] [Google Scholar]
- 191.Inglut CT, Sorrin AJ, Kuruppu T, et al. Immunological and Toxicological Considerations for the Design of Liposomes. Nanomaterials (Basel) 2020; 10(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Reeves JP, Dowben RM. Formation and properties of thin‐walled phospholipid vesicles. Journal of cellular physiology 1969; 73(1): 49–60. [DOI] [PubMed] [Google Scholar]
- 193.Pattni BS, Chupin VV, Torchilin VP. New developments in liposomal drug delivery. Chemical reviews 2015; 115(19): 10938–66. [DOI] [PubMed] [Google Scholar]
- 194.Schubert MA, Müller-Goymann CC. Solvent injection as a new approach for manufacturing lipid nanoparticles – evaluation of the method and process parameters. European Journal of Pharmaceutics and Biopharmaceutics 2003; 55(1): 125–31. [DOI] [PubMed] [Google Scholar]
- 195.Duong V-A, Nguyen T-T-L, Maeng H-J. Preparation of solid lipid nanoparticles and nanostructured lipid carriers for drug delivery and the effects of preparation parameters of solvent injection method. Molecules 2020; 25(20): 4781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Jagdevappa P, Prashant G, Ravindra K, Sachin J, Satish M, Meghanath S. Applications of solid lipid nanoparticle in novel drug delivery system. British Biomedical Bulletin 2013; 1(2): 103–18. [Google Scholar]
- 197.Zhang Y, Hu Y, Tian H, Chen X. Opportunities and Challenges for mRNA Delivery Nanoplatforms. The Journal of Physical Chemistry Letters 2022; 13(5): 1314–22. [DOI] [PubMed] [Google Scholar]
- 198.Rietwyk S, Peer D. Next-Generation Lipids in RNA Interference Therapeutics. ACS Nano 2017; 11(8): 7572–86. [DOI] [PubMed] [Google Scholar]
- 199.Oude Blenke E, Evers MJW, Baumann V, Winkler J, Storm G, Mastrobattista E. Critical evaluation of quantification methods for oligonucleotides formulated in lipid nanoparticles. International Journal of Pharmacology 2018; 548(2): 793–802. [DOI] [PubMed] [Google Scholar]
- 200.Shang L, Cheng Y, Zhao Y. Emerging droplet microfluidics. Chemical Reviews 2017; 117(12): 7964–8040. [DOI] [PubMed] [Google Scholar]
- 201.Khadke S, Roces CB, Cameron A, Devitt A, Perrie Y. Formulation and manufacturing of lymphatic targeting liposomes using microfluidics. Journal of Controlled Release 2019; 307: 211–20. [DOI] [PubMed] [Google Scholar]
- 202.Evers MJ, Kulkarni JA, van der Meel R, Cullis PR, Vader P, Schiffelers RM. State‐of‐the‐ art design and rapid‐mixing production techniques of lipid nanoparticles for nucleic acid delivery. Small Methods 2018; 2(9): 1700375. [Google Scholar]
- 203.Michelon M, Oliveira DRB, de Figueiredo Furtado G, de la Torre LG, Cunha RL. High-throughput continuous production of liposomes using hydrodynamic flow-focusing microfluidic devices. Colloids and Surfaces B: Biointerfaces 2017; 156: 349–57. [DOI] [PubMed] [Google Scholar]
- 204.Shepherd SJ, Issadore D, Mitchell MJ. Microfluidic formulation of nanoparticles for biomedical applications. Biomaterials 2021; 274: 120826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Tomeh MA, Zhao X. Recent advances in microfluidics for the preparation of drug and gene delivery systems. Molecular Pharmaceutics 2020; 17(12): 4421–34. [DOI] [PubMed] [Google Scholar]
- 206.Rosenblum D, Gutkin A, Kedmi R, et al. CRISPR-Cas9 genome editing using targeted lipid nanoparticles for cancer therapy. Science Advances 2020; 6(47). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Le-Vinh B, Steinbring C, Wibel R, Friedl JD, Bernkop-Schnürch A. Size shifting of solid lipid nanoparticle system triggered by alkaline phosphatase for site specific mucosal drug delivery. European Journal of Pharmaceutics and Biopharmaceutics 2021; 163: 109–19. [DOI] [PubMed] [Google Scholar]
- 208.Andar AU, Hood RR, Vreeland WN, DeVoe DL, Swaan PW. Microfluidic Preparation of Liposomes to Determine Particle Size Influence on Cellular Uptake Mechanisms. Pharmaceutical Research 2014; 31(2): 401–13. [DOI] [PubMed] [Google Scholar]
- 209.Petros RA, DeSimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nature reviews Drug discovery 2010; 9(8): 615–27. [DOI] [PubMed] [Google Scholar]
- 210.Stetefeld J, McKenna SA, Patel TR. Dynamic light scattering: a practical guide and applications in biomedical sciences. Biophysical Reviews 2016; 8(4): 409–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Caputo F, Clogston J, Calzolai L, Rösslein M, Prina-Mello A. Measuring particle size distribution of nanoparticle enabled medicinal products, the joint view of EUNCL and NCI-NCL. A step by step approach combining orthogonal measurements with increasing complexity. Journal of Controlled Release 2019; 299: 31–43. [DOI] [PubMed] [Google Scholar]
- 212.Filipe V, Hawe A, Jiskoot W. Critical evaluation of Nanoparticle Tracking Analysis (NTA) by NanoSight for the measurement of nanoparticles and protein aggregates. Pharmaceutical Research 2010; 27(5): 796–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Grabielle-Madelmont C, Lesieur S, Ollivon M. Characterization of loaded liposomes by size exclusion chromatography. Journal of Biochemical and Biophysical Methods 2003; 56(1): 189–217. [DOI] [PubMed] [Google Scholar]
- 214.Mohammed AR, Weston N, Coombes AGA, Fitzgerald M, Perrie Y. Liposome formulation of poorly water soluble drugs: optimisation of drug loading and ESEM analysis of stability. International Journal of Pharmaceutics 2004; 285(1): 23–34. [DOI] [PubMed] [Google Scholar]
- 215.Huang X, Chau Y. Investigating impacts of surface charge on intraocular distribution of intravitreal lipid nanoparticles. Experimental Eye Research 2019; 186: 107711. [DOI] [PubMed] [Google Scholar]
- 216.Freitas C, Müller RH. Effect of light and temperature on zeta potential and physical stability in solid lipid nanoparticle (SLN™) dispersions. International Journal of Pharmaceutics 1998; 168(2): 221–9. [Google Scholar]
- 217.Loura L, Prieto MJ. FRET in membrane biophysics: An overview. Frontiers in Physiology 2011; 2: 82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.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 Transduction and Targeted Therapy 2020; 5(1): 1-. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Chou SJ, Yang P, Ban Q, et al. Dual Supramolecular Nanoparticle Vectors Enable CRISPR/Cas9‐Mediated Knockin of Retinoschisin 1 Gene—A Potential Nonviral Therapeutic Solution for X‐Linked Juvenile Retinoschisis. Advanced Science 2020; 7(10): 1903432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Pardi N, Tuyishime S, Muramatsu H, et al. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. Journal of Controlled Release 2015; 217: 345–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Wretlind A Development of fat emulsions. Journal of Parenteral Enteral Nutrition 1981; 5(3): 230–5. [DOI] [PubMed] [Google Scholar]
- 222.Guimaraes PPG, Zhang R, Spektor R, et al. Ionizable lipid nanoparticles encapsulating barcoded mRNA for accelerated in vivo delivery screening. Journal of Controlled Release 2019; 316: 404–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Feldman RA, Fuhr R, Smolenov I, et al. mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials. Vaccine 2019; 37(25): 3326–34. [DOI] [PubMed] [Google Scholar]
- 224.Kenney RT, Frech SA, Muenz LR, Villar CP, Glenn GM. Dose sparing with intradermal injection of influenza vaccine. New England Journal of Medicine 2004; 351(22): 2295–301. [DOI] [PubMed] [Google Scholar]
- 225.Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunology, Immunotherapy 2006; 55(6): 672–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Bartesaghi S, Wallenius K, Hovdal D, et al. Subcutaneous delivery of FGF21 mRNA therapy reverses obesity, insulin resistance, and hepatic steatosis in diet-induced obese mice. Molecular Therapy - Nucleic Acids 2022; 28: 500–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Gao X, Tao Y, Lamas V, et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 2018; 553(7687): 217–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Staahl BT, Benekareddy M, Coulon-Bainier C, et al. Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nature Biotechnology 2017; 35(5): 431–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Wang H-X, Li M, Lee CM, et al. CRISPR/Cas9-based genome editing for disease modeling and therapy: challenges and opportunities for nonviral delivery. Chemical Reviews 2017; 117(15): 9874–906. [DOI] [PubMed] [Google Scholar]
- 230.Lokugamage MP, Vanover D, Beyersdorf J, et al. Optimization of lipid nanoparticles for the delivery of nebulized therapeutic mRNA to the lungs. Nature Biomedical Engineering 2021; 5(9): 1059–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Chang RYK, Chan H-K. Lipid nanoparticles for the inhalation of mRNA. Nature Biomedical Engineering 2021; 5(9): 949–50. [DOI] [PubMed] [Google Scholar]
- 232.Qin M, Du G, Sun X. Recent Advances in the Noninvasive Delivery of mRNA. Accounts of Chemical Research 2021; 54(23): 4262–71. [DOI] [PubMed] [Google Scholar]
- 233.Lobaina Mato Y Nasal route for vaccine and drug delivery: Features and current opportunities. International Journal of Pharmacology 2019; 572: 118813. [DOI] [PubMed] [Google Scholar]
- 234.Ali J, Ali M, Baboota S, et al. Potential of Nanoparticulate Drug Delivery Systems by Intranasal Administration. Current Pharmaceutical Design 2010; 16(14): 1644–53. [DOI] [PubMed] [Google Scholar]
- 235.Yadav S, Gattacceca F, Panicucci R, Amiji MM. Comparative biodistribution and pharmacokinetic analysis of cyclosporine-A in the brain upon intranasal or intravenous administration in an oil-in-water nanoemulsion formulation. Molecular Pharmaceutics 2015; 12(5): 1523–33. [DOI] [PubMed] [Google Scholar]
- 236.Dhaliwal HK, Fan Y, Kim J, Amiji MM. Intranasal delivery and transfection of mRNA therapeutics in the brain using cationic liposomes. Molecular Pharmaceutics 2020; 17(6): 1996–2005. [DOI] [PubMed] [Google Scholar]
- 237.Riley RS, Kashyap MV, Billingsley MM, et al. Ionizable lipid nanoparticles for in utero mRNA delivery. Science Advances 2021; 7(3): eaba1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Patel S, Ryals RC, Weller KK, Pennesi ME, Sahay G. Lipid nanoparticles for delivery of messenger RNA to the back of the eye. Journal of Controlled Release 2019; 303: 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Rybakova Y, Kowalski PS, Huang Y, et al. mRNA Delivery for Therapeutic Anti-HER2 Antibody Expression In Vivo. Molecular Therapy 2019; 27(8): 1415–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Nabhan JF, Wood KM, Rao VP, et al. Intrathecal delivery of frataxin mRNA encapsulated in lipid nanoparticles to dorsal root ganglia as a potential therapeutic for Friedreich’s ataxia. Scientific Reports 2016; 6(1): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Turnbull IC, Eltoukhy AA, Fish KM, et al. Myocardial Delivery of Lipidoid Nanoparticle Carrying modRNA Induces Rapid and Transient Expression. Molecular Therapy 2016; 24(1): 66–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Samaridou E, Heyes J, Lutwyche P. Lipid nanoparticles for nucleic acid delivery: Current perspectives. Advanced Drug Delivery Reviews 2020; 154–155: 37–63. [DOI] [PubMed]
- 243.Kranz LM, Diken M, Haas H, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 2016; 534(7607): 396–401. [DOI] [PubMed] [Google Scholar]
- 244.Kedmi R, Veiga N, Ramishetti S, et al. A modular platform for targeted RNAi therapeutics. Nature Nanotechnology 2018; 13(3): 214–9. [DOI] [PubMed] [Google Scholar]
- 245.Shobaki N, Sato Y, Harashima H. Mixing lipids to manipulate the ionization status of lipid nanoparticles for specific tissue targeting. International Journal of Nanomedicine 2018; 13: 8395–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T. Identification of tissue-specific microRNAs from mouse. Current Biology 2002; 12(9): 735–9. [DOI] [PubMed] [Google Scholar]
- 247.Tombácz I, Laczkó D, Shahnawaz H, et al. Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA-LNPs. Molecular Therapy 2021; 29(11): 3293–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.van de Water FM, Boerman OC, Wouterse AC, Peters JG, Russel FG, Masereeuw R. Intravenously administered short interfering RNA accumulates in the kidney and selectively suppresses gene function in renal proximal tubules. Drug Metabolism Disposition 2006; 34(8): 1393–7. [DOI] [PubMed] [Google Scholar]
- 249.Christensen J, Litherland K, Faller T, et al. Biodistribution and metabolism studies of lipid nanoparticle–formulated internally [3H]-labeled siRNA in mice. Drug Metabolism and Disposition 2014; 42(3): 431–40. [DOI] [PubMed] [Google Scholar]
- 250.Thierry AR, Rabinovich P, Peng B, Mahan LC, Bryant JL, Gallo RC. Characterization of liposome-mediated gene delivery: expression, stability and pharmacokinetics of plasmid DNA. Gene Therapy 1997; 4(3): 226–37. [DOI] [PubMed] [Google Scholar]
- 251.Mahato RI, Anwer K, Tagliaferri F, et al. Biodistribution and gene expression of lipid/plasmid complexes after systemic administration. Hum Gene Ther 1998; 9(14): 2083–99. [DOI] [PubMed] [Google Scholar]
- 252.Ishiwata H, Suzuki N, Ando S, Kikuchi H, Kitagawa T. Characteristics and biodistribution of cationic liposomes and their DNA complexes. Journal of Controlled Release 2000; 69(1): 139–48. [DOI] [PubMed] [Google Scholar]
- 253.Mullard A Novartis secures first CRISPR pharma collaborations. Nature Reviews Drug Discovery 2015; 14(2): 82–3. [Google Scholar]
- 254.Guilinger JP, Thompson DB, Liu DR. Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nature Biotechnology 2014; 32(6): 577–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Allen TM, Cullis PR. Liposomal drug delivery systems: from concept to clinical applications. Advanced Drug Delivery Reviews 2013; 65(1): 36–48. [DOI] [PubMed] [Google Scholar]
- 256.Vad BS, Bertelsen K, Johansen CH, et al. Pardaxin permeabilizes vesicles more efficiently by pore formation than by disruption. Biophysical Journal 2010; 98(4): 576–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Kim HJ, Ogura S, Otabe T, et al. Fine-tuning of hydrophobicity in amphiphilic polyaspartamide derivatives for rapid and transient expression of messenger RNA directed toward genome engineering in brain. ACS Central Science 2019; 5(11): 1866–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Luo Y-L, Xu C-F, Li H-J, et al. Macrophage-Specific in Vivo Gene Editing Using Cationic Lipid-Assisted Polymeric Nanoparticles. ACS Nano 2018; 12(2): 994–1005. [DOI] [PubMed] [Google Scholar]
- 259.Smith SA, Selby LI, Johnston AP, Such GK. The endosomal escape of nanoparticles: toward more efficient cellular delivery. Bioconjugate Chemistry 2018; 30(2): 263–72. [DOI] [PubMed] [Google Scholar]
- 260.Zangi L, Lui KO, von Gise A, et al. Modified mRNA directs the fate of heart progenitor cells and induces vascular regeneration after myocardial infarction. Nature Biotechnology 2013; 31(10): 898–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Wadhwa A, Aljabbari A, Lokras A, Foged C, Thakur A. Opportunities and challenges in the delivery of mRNA-based vaccines. Pharmaceutics 2020; 12(2): 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Cho SW, Kim S, Kim Y, et al. Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome research 2014; 24(1): 132–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Senturk S, Shirole NH, Nowak DG, et al. Rapid and tunable method to temporally control gene editing based on conditional Cas9 stabilization. Nature Communications 2017; 8(1): 14370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Lattanzi A, Meneghini V, Pavani G, et al. Optimization of CRISPR/Cas9 delivery to human hematopoietic stem and progenitor cells for therapeutic genomic rearrangements. Molecular Therapy 2019; 27(1): 137–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Liang X, Potter J, Kumar S, Ravinder N, Chesnut JD. Enhanced CRISPR/Cas9-mediated precise genome editing by improved design and delivery of gRNA, Cas9 nuclease, and donor DNA. Journal of Biotechnology 2017; 241: 136–46. [DOI] [PubMed] [Google Scholar]
- 266.Kleinstiver BP, Pattanayak V, Prew MS, et al. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016; 529(7587): 490–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Kanamala M, Wilson WR, Yang M, Palmer BD, Wu Z. Mechanisms and biomaterials in pH-responsive tumour targeted drug delivery: A review. Biomaterials 2016; 85: 152–67. [DOI] [PubMed] [Google Scholar]
- 268.Hu Z, Wang S, Zhang C, et al. A compact Cas9 ortholog from Staphylococcus Auricularis (SauriCas9) expands the DNA targeting scope. PLOS Biology 2020; 18(3): e3000686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Chu C-J, Dijkstra J, Lai M-Z, Hong K, Szoka FC. Efficiency of Cytoplasmic Delivery by pH-Sensitive Liposomes to Cells in Culture. Pharmaceutical Research 1990; 7(8): 824–34. [DOI] [PubMed] [Google Scholar]
- 270.Connor J, Huang L. pH-sensitive immunoliposomes as an efficient and target-specific carrier for antitumor drugs. Cancer research 1986; 46(7): 3431–5. [PubMed] [Google Scholar]
- 271.Ropert C Liposomes as a gene delivery system. Brazilian journal of medical and biological research 1999; 32(2). [DOI] [PubMed] [Google Scholar]
- 272.Lokugamage MP, Vanover D, Beyersdorf J, et al. Optimization of lipid nanoparticles for the delivery of nebulized therapeutic mRNA to the lungs. Nature Biomedical Engineering 2021; 5(9): 1059–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Li C, Hao M, Wang W, et al. An Efficient CRISPR/Cas9 Platform for Rapidly Generating Simultaneous Mutagenesis of Multiple Gene Homoeologs in Allotetraploid Oilseed Rape. Frontiers in Plant Science 2018; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Lyu Y, He S, Li J, et al. A photolabile semiconducting polymer nanotransducer for near‐ infrared regulation of CRISPR/Cas9 gene editing. Angewandte Chemie International Edition 2019; 58(50): 18197–201. [DOI] [PubMed] [Google Scholar]
- 275.Neefjes J, Jongsma MLM, Paul P, Bakke O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nature Reviews Immunology 2011; 11(12): 823–36. [DOI] [PubMed] [Google Scholar]
- 276.Fumoto S, Kawakami S, Hashida M, Nishida K. Targeted gene delivery: importance of administration routes. Novel Gene Therapy Approaches 2013: 3–31.
