key: cord-0907885-92tzmq97 authors: Behr, Matthew; Zhou, Jing; Xu, Bing; Zhang, Hongwei title: In vivo delivery of CRISPR-Cas9 therapeutics: Progress and challenges date: 2021-05-26 journal: Acta Pharm Sin B DOI: 10.1016/j.apsb.2021.05.020 sha: 78d2209c76e1ef90952f9f6410ebc3f9cd6054e3 doc_id: 907885 cord_uid: 92tzmq97 Within less than a decade since its inception, CRISPR-Cas9-based genome editing has been rapidly advanced to human clinical trials in multiple disease areas. Although it is highly anticipated that this revolutionary technology will bring novel therapeutic modalities to many diseases by precisely manipulating cellular DNA sequences, the low efficiency of in vivo delivery must be enhanced before its therapeutic potential can be fully realized. Here we discuss the most recent progress of in vivo delivery of CRISPR-Cas9 systems, highlight innovative viral and non-viral delivery technologies, emphasize outstanding delivery challenges, and provide the most updated perspectives. Previous genome editing technologies, such as zinc-finger nucleases and transcription activator-like effector nucleases, both of which require an individually designed protein molecule to recognize each distinct target gene, are labor intensive, costly, and time consuming. Discovery of the clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) system added a robust tool to the genome editing toolbox. It dramatically lowered the bar of conducting genome editing experiments, making it one of the most useful techniques in biomedical research 1e3 . Additionally, it has shown unprecedented potential in revolutionizing treatment of human diseases 4e6 . CRISPR-Cas9 therapeutics hold apparent advantages over traditional gene therapies, such as gene replacement therapy, RNA interference (RNAi) therapy, and antisense therapy, in that the repair on a mutated gene or the insertion of a missing gene is permanently and precisely ironed to the host genome after a single genome editing event. Due to its high accuracy of target gene recognition, CRISPR-Cas9 averts activation of oncogenes and mutagenesis. In addition, it eliminates repeated dosing in certain applications, although repeated administration of the CRISPR-Cas9 system has improved genome editing efficiency in some preclinical studies. CRISPR-Cas9 therapeutics remind everyone with ever stronger signal that the era of precision medicine is coming. Although some diseases are very likely to quickly benefit from CRISPR-Cas9 technology through ex vivo manner, the more profound clinical success of CRISPR-Cas9 therapeutics can only be accomplished if the system can be directly administered to patients. However, safe and effective in vivo delivery systems for the components of CRISPR-Cas9 therapeutics are still yet to mature (Fig. 1 ). This article summarizes the most recent progress of in vivo delivery of CRISPR-Cas9 systems for genome editing, highlights innovative delivery technologies using viral and nonviral vectors, emphasizes outstanding challenges to in vivo delivery faced by the CRISPR-Cas9 genome editing community, and provides the most updated perspectives. Though CRISPR-Cas systems have bacterial origins from species such as, but not limited to, Streptococcus pyogenes, Staphylococcus aureus, and Streptococcus thermophilus, their utility reaches beyond the bacterial immune system, and can be used for genome editing in mammalian cells 2, 7 . CRISPR-Cas systems can be differentiated based on the number of Cas proteins involved in their mechanism. A type I system uses multiple Cas proteins while a type II system uses only one Cas protein 8, 9 . Because of this contrast, type II CRISPR-Cas systems are more desirable for genome engineering as its components can be produced with greater ease. Type II CRISPR-Cas systems involve multiple key components for genome engineering, including a Cas protein, CRISPR RNA (crRNA), and transactivating crRNA (tracrRNA) 8,10e12 . Cas9 is an endonuclease with two nuclease domains. These domains are the HNH, which cleaves complementary DNA strands, and RuvC, which cleaves noncomplementary DNA strands 2, 8 . Mammalian genome editing with CRISPR-Cas9 was simplified by engineering a single guide RNA (sgRNA), which takes the place of crRNA and tracrRNA components in the CRISPR-Cas system 13 . The sgRNA is essential for precise gene editing as its 5ʹterminal 20-nucleotide sequence interacts with a target sequence of host DNA via Watson and Crick base pairing rules, while its 3ʹ duplex structure allows binding to Cas9 proteins 8 . Before being able to perform a double strand break (DSB), Cas9 proteins must locate the target DNA sequence by recognizing the protospacer adjacent motif (PAM), which is a short sequence of nucleotides directly adjacent to the target DNA sequence 8, 11, 14 . Once a Cas9 protein binds to the target strand of DNA, it undergoes a conformational change, allowing for a targeted DSB to occur 3e4 nucleotides upstream of the PAM 8, 11, 15 . Following a DSB, there are multiple fates for the broken chromosomal DNA. Primary DSB repair mechanisms include nonhomologous end joining (NHEJ) or homology directed repair (HDR) pathways (Fig. 2) . NHEJ involves direct ligation of the two broken chromosomal DNA strands to one another and is the main cellular DSB repair mechanism. This process is error-prone due to random insertions or deletions (indels) of nucleotides to assist in ligation, which can cause gene disruption via frameshift or nonsense mutations 16 . Beyond gene disruption, NHEJ can also be used to induce significant deletions when carried out with two different sgRNAs. Gene disruption or deletion via NHEJ is desirable when treating diseases whose pathogenesis is characterized by an over-expression of a certain protein, such as transthyretin amyloidosis. HDR pathways require the presence of an exogenous donor DNA template that is homologous to the target region of chromosomal DNA, allowing for gene insertion following DSB. HDR's applications rest in targeted integration of transgene. Gene editing with HDR is the desired manner for the treatment of diseases caused by missing an entire gene, such as X-linked retinitis pigmentosa, hemophilia A or B, and phenylketonuria. When compared, NHEJ is more efficient and can occur at any phase of the cell cycle. It is utilized to edit DNA more frequently than HDR, which is less efficient, especially in post-mitotic cells, such as neurons, retinal pigment epithelial (RPE) cells, cardiac myocytes and mature muscle cells. These two repair mechanisms are most used by researchers as they allow for a straightforward approach to gene disruption, deletion, correction, or insertion. Alternative DSB repair mechanisms include homologymediated end joining (HMEJ), microhomology-mediated end joining (MMEJ), and homology-independent targeted integration (HITI). MMEJ's mechanism includes 5e25 bp microhomologous Figure 1 Milestones of in vivo delivery of CRISPR-Cas9 therapeutics. Key developmental milestones for in vivo delivery of CRISPR-Cas9. AAV: adeno-associated virus, NP: nanoparticle, LNP: lipid nanoparticle. CRISPR: clustered regularly interspaced short palindromic repeats, Cas9: CRISPR-associated protein 9. arm sequences that align between both strands of the broken DNA, allowing for end joining once the nucleotide sequences directly adjacent to the original break and microhomologies are cleaved 17, 18 . MMEJ can be utilized as a mechanism for gene insertion that requires shorter homology arms than HDR 18, 19 . Additionally, HMEJ works in a similar mechanism to MMEJ, but instead with 800 bp homologous arm sequences to assist with DNA repair or transgene insertion 19 . Lastly, HITI requires administration of an exogenous donor DNA template for insertion, however unlike HDR, HITI does not require chromosomal DNA homology for insertion, and instead utilizes a three sgRNA system to cleave chromosomal and donor DNA prior to insertion 20 . Although Cas9 protein, sgRNA, and donor DNA template (if for HDR) must present at the nucleus of target cells for genome editing therapy, they are not necessarily the cargo originally loaded into the delivery systems for administration. To enable CRISPR-Cas9-based genome editing in vivo, there are multiple choices in terms of cargoes. Direct delivery of Cas9/sgRNA ribonucleoprotein (RNP) complexes is the most straightforward choice, due to the high genome editing efficiency, rapid onset of action, short resident time in the target cell, and reduced off-target Figure 2 CRISPR-Cas9 genome editing through viral or non-viral delivery. Representative depiction of mechanisms and strategies involved in CRISPR-Cas9 delivery with both viral and non-viral vectors. AAV and lentivirus both bind to cell surface receptors prior to cellular infection. Following cellular internalization, AAVs have the capacity to escape the endosomes and transport across the nuclear membrane prior to uncoating, though the capsid degradation mediated by proteasome can also occur in the cytoplasm. Following lentiviral cell membrane fusion is uncoating and release of its RNA contents, which then undergo reverse transcription to form complementary DNA. Non-viral vectors offer the advantage of carrying various forms of CRISPR-Cas9 cargoes including plasmid DNA, RNA, donor DNA, and RNP. Cellular entry of non-viral vectors is accomplished via endocytosis which requires the NP to escape these endosomes in order to carry out its intended genome editing. Following endosomal escape and cytosolic release, the cargo carried by a non-viral NP must travel to varying sites, such as the nucleus for transcription and/ or cytoplasm for translation. Once necessary transcription and translation steps have taken place with nucleic acid delivery approaches, a RNP is formed and can translocate across the nuclear membrane for targeted genome editing. RNPs work to perform targeted DSBs by PAM-and sgRNA-mediated recognition of a specific sequence of chromosomal DNA. Once this recognition occurs, the Cas9 nuclease can perform a DSB utilizing its two nuclease domains the HNH and RuvC which cleave complementary and non-complementary DNA strands, respectively. Following a DSB, there are multiple fates for genome editing such as, but not limited to, NHEJ and HDR. NHEJ is utilized for genomic disruption or deletion, while HDR is utilized for gene correction, but requires the administration of an exogenous donor DNA template. AAV: adenoassociated virus, NP: nanoparticle, CRISPR: clustered regularly interspaced short palindromic repeats, Cas9: CRISPR-associated protein 9, RNP: ribonucleoprotein complexes, PAM: protospacer adjacent motif, DSB: double-stranded break, NHEJ: non-homologous end joining, HDR: homology-directed repair. effects. Alternatively, both the Cas9 protein and sgRNA may be replaced by other forms of entities, such as plasmid DNA, mRNA, or a viral genome inside a viral vector. In this case, Cas9 proteins and sgRNAs are produced on site in the target cells to form RNPs for genome editing. In addition, when HDR is the desired approach, even the donor DNA sequence can be delivered by way of plasmid DNA or viral genome. Depending on the disease, desired dosing regimen, route of administration, and delivery system, researchers have a variety of options to deliver DNA, RNA and/or protein payloads to the target cells (Fig. 2 ). There are four options for Cas9 enzymes: protein, plasmid DNA, mRNA, or viral genome, three options for sgRNA: RNA, plasmid DNA, or viral genome, and three options for DNA donor template: DNA oligos, plasmid DNA, or viral genome. Mathematically, there are twelve options (by multiplying four by 3) for NHEJ-mediated genome disruption or deletion, and thirty-six options (by multiplying four by three by 3) for HDR-mediated gene insertion. In this section, we discuss the options available for each component, analyze advantages and disadvantages of the options, and provide general guidance for consideration when making selections. Purified Streptococcus pyogenes Cas9 (SpCas9) protein is a positively charged molecule with a molecular weight of w163 kDa. The large molecule size represents a problem for efficient entry into both the cell and cell nucleus. In addition, its bacterial origin makes the Cas9 protein immunogenic in humans, which may decrease genome editing efficiency and increase the risk of a genome editing therapy 21 . Pre-existing anti-Cas9 antibodies have been detected in humans due to the frequent interaction with bacteria in the past thousands of years 22 . Furthermore, the purification process is still costly and time consuming. Moreover, it requires significant work to ensure the purity, and maintain stability and biological activity of Cas9 protein during the entire purification, formulation, shipping, and storage process. Using mRNA or plasmid DNA encoding Cas9 can avoid problems related to protein production and purification while reducing the cost. If mRNA or plasmid DNA are successfully delivered, Cas9 can be produced in target cells where gene manipulation is expected to occur. However, intracellular delivery is difficult for both naked mRNA and plasmid DNA as they are unstable and unable to enter target cells by themselves. When a comparison is made between them, mRNAs are more fragile and expensive. However, mRNAs do not pose the risk of insertional mutagenesis on chromosomal DNA. What's more, mRNAs do not need to enter cell nucleus and can produce Cas9 protein at cytosolic ribosomal sites as quickly as w6 h post intracellular delivery. This is a significant advantage over plasmid DNA which must be delivered into the cell nucleus to initiate Cas9 expression. In addition, mRNA-mediated Cas9 expression is more transient compared to plasmid DNA-mediated Cas9 transgene expression in target cells, further minimizing the exposure of the cellular genome to Cas9 and reducing the risk of off-target effects. The fourth option to deliver Cas9 is viral gene delivery. Cas9 genes can be engineered into the recombinant genome of viral vectors, such as adeno-associated viruses (AAVs) or lentiviruses, which are proficient in producing Cas9 in target cells. However, it has been shown that long-term Cas9 production in target cells led to increased off-target effects and immunogenicity 21, 23, 24 . Viral vectors also pose the risk of random gene integration into the host genome. Although both SpCas9 and Staphylococcus aureus Cas9 (SaCas9) genes can be delivered by AAVs, it is important to note that the genes of some novel Cas nucleases are too long to be packaged in a single AAV vectors. Finally, mRNA, plasmid DNA, and viral genome all require additional time to produce Cas9 enzymes in target cells, resulting in delayed action. The delay may decrease genome editing efficiency when an sgRNA is delivered concurrently, because sgRNAs may be degraded in the cell before Cas9 protein is made. An sgRNA is a negatively charged macromolecule with a molecular weight of w31 kDa. Due to the ease of synthesis, sgRNA is a popular cargo choice. Alternatively, plasmid DNA and viral genome offer appealing advantages by expressing sgRNA right in target cells. Plasmid DNA takes additional time to transcribe sgRNA in the cell, causing a delayed expression of sgRNA. If Cas9 nuclease is co-delivered along with plasmid DNA encoding sgRNA, Cas9 nucleases may not survive in the cell by the time when sgRNA is expressed. This timing issue will lead to a decreased genome editing efficiency. However, if a plasmid DNA is delivered to express both Cas9 and sgRNA in target cells, the timing issue is generally not a concern. If sgRNA is expressed by viral vectors such as AAVs, sgRNA may be produced in target cells for a prolonged period of time. This may provide convenience and flexibility for Cas9 delivery as Cas9 protein is short-lived in cells. The selection of a donor DNA template applies to HDR-based and HITI-based genome editing. Exogenous DNA template can be delivered in the form of plasmid DNA, usually when the insertion is a long DNA sequence. Alternatively, the DNA donor can be delivered as a synthetic single-stranded DNA oligonucleotide when the insertion sequence is short. Although a plasmid DNA is more stable than a short DNA oligonucleotide, its intranuclear delivery efficiency is lower due to the larger size. Moreover, when a plasmid DNA is delivered, it is important to make sure that there is no CRISPR targeting site in the plasmid DNA. Otherwise, the HDR-edited gene locus will be cut by Cas9 protein, resulting in a failed chromosomal DNA repair. AAV vectors can also deliver the DNA donor template 25e27 , or simultaneously deliver both Cas9 and the donor DNA 28 . In the latter case, it helps increase gene editing efficiency because a single AAV vector gets two genome editing components into the same target cell. CRISPR-Cas9 system can be efficiently delivered into mammalian cells by different ways such as using delivery vectors or physical methods. Delivery vectors will be discussed in depth in this section (Table 1) . However, physical methods, including microinjection and electroporation, are more applicable to ex vivo CRISPR-Cas9 based genome editing therapy. Thus, they are not discussed in this review. We suggest interested readers refer to the cited review articles covering physical methods 29e32 . Viruses are natural experts for in vivo gene delivery 33 . Recombinant viral vectors have been in leading positions for human gene 41 . Clinical success of AAV-based gene therapies has been well illustrated by regulatory approvals of Zolgensmaâ, Luxturnaâ, and Glyberaâ 36 . Though AAVs represent an excellent platform for gene therapy, they still possess weaknesses when used for in vivo delivery of CRISPR-Cas9. One significant drawback is the packaging limitation, which complicates all-in-one delivery as SpCas9 itself is about 4.2 kb in size. Since recombinant AAVs must also contain regulatory elements essential for gene expression, such as the promoter region and polyadenylation signal, the small packaging capacity creates significant inconvenience and disqualifies AAVs for many applications where large gene sequences must be delivered. Solutions to the packaging limit challenge include using a truncated SpCas9, which is smaller in size but offers reduced genome editing efficiencies, or using SaCas9, which is about 3.1 kb in size and shows comparable genome editing efficiencies 42, 43 . An alternative method is a dual-AAV strategy that splits the components of the transgene cassette into two independent viruses that are then co-infected 44, 45 . Utilizing dual AAVs is especially valuable for HDR-based gene correction and insertion as a donor DNA template must also be included with Cas9 and sgRNA. Another significant problem faced by AAVs is the pre-existing neutralizing antibodies in patients from prior AAV infection, which requires serological testing before one can be treated 38 . AAV's long-term transgene expression can be seen as a merit for sustained gene therapy, however, it can also be a risk as continuous expression of Cas9 nucleases may cause significant off-target effects 46 . Lastly, large-scale production of AAV is still difficult 47e49 . Since the first report of AAV-based CRISPR-Cas9 delivery for in vivo genome editing, it has made exciting progress in numerous disease models, including blood disorders 50e52 , metabolic liver diseases 44,53e55 , muscular diseases 56e58 , and ocular diseases 59 Lentivirus is an enveloped RNA virus with a single stranded genome that belongs to the retrovirus family 61e65 . Unlike other retroviruses, lentiviruses and can infect both dividing and nondividing cells. Lentiviruses, such as HIV-1, are more complex than retroviruses as they contain an additional six genes; tat, rev, vpr, vpu, nef, and vif that are involved in viral replication and pathogenesis. However, some of these accessory genes involved in the virulence of lentiviruses can be removed to decrease their immunogenicity 66, 67 . Typically, lentiviruses integrate their genomes into the host genome, which can significantly prolong transgene expression time, but this may increase the risk for adverse effects. Researchers have modified lentiviral integrase DNA to induce mutations leading to prevention of proviral integration, forming what is known as integration defective lentiviruses (IDLV) 71 . Benefits of using lentiviruses for in vivo gene therapy include its 10 kb carrying capacity and prolonged transgene expression 66, 68 . These attributes allow for decreased challenges with packaging limit when compared with AAV. Disadvantages of in vivo utilization include the potential for oncogenicity, insertional mutagenesis, and supra-physiological levels of RNA and/or proteins due to prolonged transgene expression time, however, these are reduced when using IDLVs 69 . Additionally, the inclusion of multiple regulatory genes involved in replication may further complicate the packaging construct 69 . Though they have promise as an in vivo delivery system, lentiviral vectors are an excellent option for ex vivo gene editing as they offer efficient cellular transduction in numerous cell types and shorter culture times needed for cell transfection 70 . These attributes make ex vivo editing with lentiviruses desirable due to their relative ease of use, which lessens challenges associated with large scale production needed for clinical utility. Preclinical studies have demonstrated the utility of lentiviral vectors for both systemic and localized in vivo delivery of Cas9 and sgRNA for therapeutic gene disruption in wet age-related macular degeneration (AMD) and cancer models 71e73 . When used for ex vivo transduction, lentivirus has proved superior to other vectors in clinical studies. Two lentiviral vector-based ex vivo gene therapeutics, Kymriahâ and Zyntegloâ, have been approved for use by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA), respectively 36 . Many types of non-viral vectors used for protein, gene and RNAi delivery were adopted or modified for in vivo CRISPR-Cas9 delivery. These include liposomes, lipid nanoparticles (LNPs), polymeric nanoparticles, peptide nanoparticles, and inorganic nanoparticles (Fig. 4) . They possess multiple attractive advantages, such as minimal immunogenicity, low cost in large scale production, the capability to deliver all components of a CRISPR-Cas9 system within one vector (all-in-one delivery), and the flexibility to deliver various cargoes (RNP, plasmid DNA, mRNA, sgRNA, donor DNA) 32, 74 . Multiple non-viral vectors are evaluated for delivery of genes or RNAi in clinical studies. Liposomes have been used for drug delivery for multiple decades 75 . Liposomes are characterized by a hydrophobic lipid bilayer and an inner aqueous pool 76 . In recent years, LNPs joined liposomes to address the challenge of safe and efficient gene delivery. LNPs are structurally distinct from liposomes, as the continuous lipid bilayer and large inner aqueous pool are absent 77 . However, both types of nanocarriers are primarily composed of natural phospholipids or their derivatives, cholesterol, cationic or ionizable lipids, and polyethylene glycol (PEG)-lipids. To condense and protect the negatively charged DNA or RNA cargoes, permanently charged cationic lipids, such as 1,2-di-Ooctadecenyl-3-trimethylammonium propane (DOTMA), 1,2dioleoyl-3-trimethylammonium-propane (DOTAP) and dimethyldioctadecylammonium (DDAB), were commonly used in early versions of the nanoparticles for the delivery of plasmid DNA and RNA molecules (e.g., siRNAs, miRNAs and mRNAs). In the past decade or so, these permanently charged lipids were gradually surpassed and replaced by ionizable cationic lipids, such as ALC-0315 and SM-102, which demonstrated superior transfection efficiency and reduced cytotoxicity for mRNA delivery 78 . With the surge of preclinical and clinical studies of genome editing therapies, a great amount of knowledge accumulated for DNA and RNA delivery was quickly translated into the delivery of CRISPR-Cas9 79 . Although cationic liposomes were the very first type of non-viral vector for traditional human gene replacement therapy 80, 81 , their application in gene therapy is still in the clinical investigation stage. In comparison, LNP-based Onpattroâ has already received regulatory approval. Very recently, two LNP-based mRNA vaccines were authorized for emergency use in the United States to fight against COVID-19 pandemic 82, 83 . LNPs have been used in many preclinical studies for CRISPR-Cas9 delivery in the past several years 84e89 . The most promising application is in the genome editing of liver cells. Several groups reported that upon intravenous administration, LNPs efficiently deliver the payloads to liver cells due to the strong liver tropism induced by low-density lipoprotein (LDL) receptors on liver cells 77, 86, 90, 91 . Recently, Siegwart and colleagues reported a novel modifiable LNP platform called selective organ targeting (SORT) LNP, by adding a fifth lipid component to the established LNP formulation. SORT LNPs delivered different CRISPR cargoes, including RNPs and Cas9 mRNA/sgRNAs, for efficient genome editing in the liver, lungs, and spleen after intravenous administration 92 . Although this strategy is very promising to expand the usefulness of LNPs, the most possible reason why SORT LNPs preferentially accumulated in the liver and spleen is because they are easily opsonized and captured by the reticuloendothelial systems. In addition, selective delivery to the lungs may result from the positive surface charge of intravascular SORT LNPs. Therefore, to achieve more success in other tissues, such as the brain, heart and kidney, stealth SORT LNPs will be needed. Nonetheless, LNP-delivered CRISPR-Cas9 therapeutics has made rapid progress. In 2020, Intellia Therapeutics' NTLA-2001, a CRISPR-Cas9 gene editing system delivered by LNP, was injected into a patient in a human clinical trial (NCT04601051) for the treatment of transthyretin amyloidosis. Polymer nanoparticles have gained popularity as delivery systems due to their numerous strengths including controlled drug release, payload protection, minimal immunogenicity, biodegradability, and improved bioavailability of active components. Polymerbased delivery vectors have been used in clinic for small molecular weight drugs and protein/peptide drugs for many years. The safety and effectiveness of polymeric nanoparticles in gene therapy have been investigated in multiple clinical trials 93 . Commonly used polymers for gene and protein delivery include poly (lacticco-glycolic-acid) (PLGA), poly (b-amino ester) (PBAE), polyethylenimine (PEI), cyclodextrin (CD) derivatives, and poly (ethylene-glycol) (PEG). Though these nanoparticles hold great promise as delivery systems, they also provide the challenge of variable biocompatibility and toxicity. Polymer nanoparticles have shown utility as in vivo delivery systems for CRISPR-Cas9 payloads. Chen et al. 94 coated preassembled RNPs by a thin glutathione-cleavable covalently crosslinked polymer layer and yielded a very fine nanocapsule with a diameter of only w25 nm. In an Ai14 mouse strain, this polymeric nanocapsule induced efficient genome editing in retinal pigment epithelium cells following subretinal injection, and skeletal muscle after intramuscular injection. In another study, Wan et al. 95 skillfully leveraged the amides and oxyanions that specifically present in Cas9 proteins and complexed RNPs with low-molecular-weight PEI modified CD through hydrogen bonding and salt bridge effects. The resulting stable nanoparticle efficiently enabled genomic disruption of mutant KRAS in a murine tumor model. Other polymer-based or hybrid delivery systems have also shown success in delivering CRISPR-Cas9 components in preclinical studies, including solid-tumor malignancies 96e98 , and disease states characterized by over-activation of the immune system, such as septic shock 99 , peritonitis 99 , allograft rejection 100 , glioma 101 , and type two diabetes mellitus 99,100,102,103 . Peptides have attracted much attention in CRISPR-Cas9 delivery because they can be used as not only backbone materials, but also surface modifying molecules of nanoparticles. Several types of peptides have been explored for self-assembly of nanoparticles. Peptide nanoparticles aid in payload protection, cellular targeting, intracellular delivery, and endosomal escape 104e106 . Though peptide-based delivery mechanisms have multiple positive aspects, they are still faced with multiple challenges, such as immunogenicity, reduced activity due to protease degradation, and potential toxicities caused by foreign peptides. Peptide nanoparticles and conjugates have been used successfully in delivering CRISPR-Cas9 components in preclinical studies for genome editing 107e111 . Recently, He and colleagues reported a novel self-assembled peptide nanoparticle which selectively delivered CRISPR-Cas9 components to the mitochondria of cancer cells for mitochondrial DNA editing 112 . This work greatly expanded the potential applications of CRISPR-Cas9 to mitochondrial DNA editing. Though these preclinical models shown positive therapeutic effects, peptide-based delivery systems have yet to enter clinical trials involving CRISPR-Cas9 therapeutics. Gold nanoparticles have been used to aid in the delivery of imaging agents, nucleic acids and proteins. Gold is an attractive option for nanoparticle formulation as it is relatively biocompatible, can permeate multiple cell types, and can be conjugated with other components, such as cell penetrating peptides, nucleic acids, lipids and/or polymers to increase delivery efficiency 113, 114 . Although these nanoparticles are considered relatively non-immunogenic, their toxicity profile is largely dependent on physicochemical properties such as size, shape, charge, and surface modifications 115, 116 . Recently, gold nanoparticles have been used in preclinical studies as a medium for conjugation of Cas9-sgRNA RNPs or Cas9 protein and sgRNA-encoding plasmids for in vivo genome editing via local administration 117, 118 . Silica nanoparticles are another noteworthy delivery system, which have shown the capability to successfully delivery CRISPR-Cas9 to multiple organs and cell types for genome editing. These delivery systems have seen an introduction into CRISPR delivery as they are very versatile, biocompatible, and can be modified in multiple ways based on delivery strategy 119 . Some of this versatility is seen in alterations of particle size as well as chemical modifications which allow for better encapsulation and/or conjugation of CRISPR-Cas9 components or other materials involved in delivery, such as lipids or polymers. When utilized for CRISPR delivery in preclinical studies, these nanoparticles are commonly mesoporous and formulated with tetraethyl orthosilicate (TEOS) as well as conjugated with some form of lipid or organic compound. Multiple preclinical applications demonstrated the potential of silica nanoparticles for in vivo delivery of CRISPR-Cas9 to treat murine models of hypercholesterolemia 120 , hepatocellular carcinoma 121 , pulmonary adenocarcinoma 122 , and melanoma 123 as well as proof-of-concept editing in retinal pigment epithelium 124 and brain neurons 125 . Many diseases require safe and efficient in vivo delivery of CRISPR-Cas9 components in order to enable accurate genome editing in target cells and tissues. So far, multiple viral vectors and non-viral vectors have demonstrated the usefulness for CRISPR-Cas9mediated genome editing in various tissues in preclinical studies. The liver is a popular target for therapeutic gene editing as it involves in metabolism and the production of numerous essential proteins. CRISPR-Cas9 has been administered systemically with both viral and non-viral vectors for therapeutic gene editing in the liver to treat conditions such as hemophilia, familial hypercholesterolemia, phenylketonuria, and ornithine transcarbamylase deficiency. Hemophilia B is an X-linked condition that results from dysfunction or deficiency of coagulation factor IX (FIX). Recently, Wang et al. 52 utilized a dual AAV8 delivery system to intravenously deliver SpCas9 and sgRNA genomes as well as codon-optimized partial human FIX for HDR at the albumin locus in hepatocytes of hemophilia B-induced mice 52 . CRISPR-Cas9 treated adult and neonatal hemophilia B-induced mice showed a site-specific gene integration with an on-target indel percentage of 21%e34% in adults and 26.1%e40.1% in neonates as well as an HDR percentage of 3.0%e4.6% in adults and 8.2%e16.1% in neonates. Both adult and neonatal mice exhibited a significant increase in FIX protein and FIX activity when compared to control treated mice, and these results were sustained until the study was completed 48 weeks post-injection (Fig. 3A) . Familial hypercholesterolemia can be related to overexpression of proprotein convertase subtilisin kexin 9 (PCSK9), as it regulates the degradation of LDL receptors in hepatocytes in response to blood cholesterol levels 126 . This disease is inherited in an autosomal dominant pattern and leads to increased levels of LDL in the bloodstream, causing atherosclerosis and thromboembolic events 127 . Liu et al. 88 utilized bioreducible LNPs formulated with cholesterol, DOPE, and DSPE-PEG2000 to deliver Cas9 mRNA and sgRNA targeting PCSK9 for NHEJ-mediated disruption in mice hepatocytes via single intravenous injection. CRISPR-Cas9 therapy led to a 20% decrease in serum PCSK9 when compared to untreated mice. Treated mice also showed no significant changes in aspartate aminotransferase (AST), alanine aminotransferase (ALT), and total bilirubin when compared to mice treated with phosphate buffered saline. Phenylketonuria is a metabolic liver disease inherited in an autosomal recessive manner in which the body has a deficiency of the enzyme phenylalanine hydroxylase (PAH), leading to decreased metabolism of L-phenylalanine and hyperphenylalaninemia 128 . Those untreated with this condition develop neurologic complications due to hyperphenylalaninemia. Villiger et al. 54 demonstrated effective administration of a dual AAV8 delivery system via intravenous injection to phenylketonuriainduced mice. AAVs with genomes encoding a SaCas9 base editor and a sgRNA targeting PAH in hepatocytes induced an ontarget indel percentage of up to 10.5% and gene correction of up to 63% in hepatocytes. PAH correction led to a significant increase in PAH enzyme activity, a decrease in blood phenylalanine levels, and maintenance of mouse body weight and fur pigmentation. Following AAV8 therapy, mice showed no significant off-target indels at ten predicted loci as well as no indication of DNA damage or cell proliferation. Ornithine transcarbamylase deficiency (OTCD) is inherited in an X-linked recessive manner and affects key components of the urea cycle causing supra-physiological levels of ammonia to accumulate, which can lead to neurological complications and mortality 129, 130 . Wang et al. 55 showed the utility of gene correction with dual AAV8 delivery of genomes expressing SaCas9, sgRNA targeting OTC in hepatocytes, and donor DNA template via intravenous injection for the treatment of OTCD in mice. CRISPR-Cas9 therapy induced an on-target indel percentage that ranged from 22% to 38% and an HDR efficiency of 2%e10%. Genome editing effects were seen at three and eight weeks when liver samples were harvested for analysis. CRISPRtreated mice showed restoration of OTC activity up to 79% of wild type as well as significantly increased survival time and lower levels of ammonia, which were also comparable to levels in wild type mice. These studies showed encouraging results for therapeutic genome editing in the liver. Studies conducted by both Wang et al. 52 and Villiger et al. 54 demonstrated the ability for prolonged therapeutic effects of up to 48 and 26 weeks, respectively. Alternatively, the studies conducted by Liu et al. 88 and Wang et al. 55 lacked off-target indel data and a long-term follow-up period, leaving the long-term efficacy and toxicity of each therapy to be further investigated. CRISPR-Cas9 can serve as a novel treatment method for various malignancies as NHEJ-mediated gene disruption or deletion is a significant tool in blocking tumor growth mechanisms and/or immune resistance. Systemic injection is typically the preferred route of administration because many cancers are metastatic and present with tumor involvement in multiple organs. However, CRISPR-Cas9 components can also be delivered via intratumoral injection to treat localized cancer. CRISPR-Cas9 therapeutics have showed success as a treatment in preclinical malignancy models, including triple negative breast cancer, nasopharyngeal carcinoma, and melanoma. Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer that is highly metastatic. Guo et al. 97 have demonstrated the ability of a tumor targeting nanolipogel (tNLG) delivery system for deleting oncogene lipocalin 2 (LCN2). The tNLG delivery system is a non-cationic, lipidepolymer hybrid, composed of DOPC, and DSPE-PEG-COOH as well as alginate. TNLGs are covalently conjugated with an antibody targeting intracellular adhesion molecule 1 (ICAM1) to maximize editing efficiency and specificity. This robust delivery system encapsulated three plasmids expressing Cas9 and sgRNAs targeting differing DNA sequences of LCN2 for NHEJ-mediated deletion and was administered via repeated intravenous injections. TNLG therapy induced an on-target genome editing efficiency of 80%, which led to significant decreases in LCN2 expression as well as tumor volume and mass when compared to control-treated mice. Additionally, this therapy was only associated with minimal systemic toxicity. CRISPR-Cas9 genome editing therapy was also combined with photodynamic therapy for cancer treatment. Deng et al. 131 codelivered CRISPR-Cas9 RNP with photosensitizer chlorin e6 to a xenograft nasopharyngeal carcinoma mouse model using a near infrared (NIR) light and reducing environment-responsive polymeric nanoparticle. This delivery system was administered via repeated intravenous injection and encapsulated SpCas9/sgRNA RNP targeting nuclear factor erythroid 2-related factor 2 (NRF2), an antioxidant regulator, for NHEJ-mediated disruption in CNE2 tumor cells. Upon administration, NIR light was exposed to the tumor location, which destabilized lysosomal membranes and allowed the entry of Cas9/sgRNA RNP into the cytoplasm. This therapy induced an on-target indel percentage of 26.7%e32.28% with off-target percentages up to 4.1% in the tumor and liver tissues. Treatment with Cas9/sgRNA, chlorin e6, and NIR (T-CC/ NIR) led to a decreased expression of NRF2, hypoxia-inducible factor 1a (HIF1a), and vascular endothelial growth factor-A (VEGF-A). Additionally, T-CC/NIR-treated mice showed an increased survival time by 80% as well as significantly decreasing tumor volume when compared to controls. CRISPR-treated mice had similar tissue structure in major organs and body weight fluctuations when compared to untreated mice. The over-expression of cyclin dependent kinase 5 (CDK5) in melanoma and TNBC is involved in angiogenesis, apoptosis, and immune checkpoint regulation of tumor cells 132 . In a recent study, Deng et al. 98 used a poly (beta-amino-ester) copolymer nanoparticle to administer a SpCas9/sgRNA expressing plasmid targeting CDK5 for NHEJ-mediated disruption. CDK5 disruption has exhibited the ability to decrease levels of programmed-death ligand-1 (PD-L1) which activates its receptor, programmed cell death protein (PD-1), on T cells, leading to decreased T cell activation and increased tumor cell proliferation 133, 134 . In both models, CRISPR therapy led to significant reductions in the expression of CDK5, PD-L1, and P35, as well as decreased tumor volume, weight, and lung metastasis (TNBC model). CDK5 disruption led to greater inhibition of tumor growth than the administration of anti-PD-L1 antibodies, and in the melanoma model, prolonged lifespan to 40 days compared to 26 days in PBStreated mice. CRISPR-treated mice experienced no severe systemic toxicity (Fig. 4) . None of the preclinical malignancy studies reported long-term data after genome editing treatment, which is most likely due to the severity and progression of disease states. Also, some studies lacked indel data which complicates conclusions on their potential efficacy and safety. Interestingly, from antibody conjugation, to immune checkpoint inhibition, to NIR exposure, each study utilized unique tactics to improve genome editing effects and specificity. Creative approaches such as these may prove to be a key component for the future clinical application of in vivo CRISPR therapies. Skeletal and cardiac muscle have been targeted for CRISPR-based gene editing using both systemic and local injection strategies. Treatment with both non-viral and viral delivery systems has shown promise in conditions such as DMD and catecholaminergic polymorphic ventricular tachycardia (CPVT). DMD is a disease typically inherited in an X-linked recessive manner resulting from mutations in the DMD gene that encodes the protein dystrophin 5 . Loss-of-function mutations in DMD causes muscle weakness and wasting that can be fatal as dystrophin plays a crucial role in muscle structure and function by stabilizing the sarcolemma 135 . Zhang et al. 56 reported successful rescue of DMD mice through in vivo genome editing by using a dual AAV delivery strategy, with ssAAV-packaged SpCas9 and ssAAV-or scAAV-packaged sgRNA. In mice with exon 44 of DMD deleted, intraperitoneal injection of AAV9 targeting exon 45 of DMD in skeletal and cardiac myocytes resulted in exon skipping or reframing therapy and caused dose-dependent on-target indel percentages of up to 30%. On-target indels were significantly higher in tibialis anterior (TA) and triceps when treated with scAAVs package sgRNA. CRISPR-treated mice experienced significant restoration of wild type dystrophin in TA, triceps, diaphragm, and heart. Treated mice also showed a significant increase in specific force from extensor digitorum longus and soleus muscles, as well as increased maximal tetanic force when compared to untreated mice, with scAAVs more so than ssAAVs. Genome editing effects persisted for 4 weeks following AAV delivery when mice were sacrificed for analysis. CRISPR-Cas9 mediated genome editing in muscular tissues was also achieved using extracellular vesicles (EVs). Gee et al. 136 loaded RNP into an extracellular nanovesicle called NanoMEDIC (nanomembrane-derived extracellular vesicles for the delivery of macromolecular cargo), and demonstrated successful genome editing in cultured cells and in mdx mice. Efficient loading of RNP into EVs in producer cells was achieved through a chemicalinduced dimerization process. In addition, an expression vector containing two lentiviral vector elements, including HIV-1 Tat, was used to express sgRNA. Moreover, self-cleaving ribozymes were flanked to sgRNA to facilitate its release in target cells. Despite the elegant yet complicated design, NanoMEDIC encapsulated SpCas9/sgRNA RNPs enabled exon skipping in muscular cells in a luciferase mouse model for up to 160 days, indicating the potential for in vivo DMD treatment. Although this proof-ofconcept study did not report off-target editing or adverse event data, Tat may be included in EVs, which represents a potential risk factor to targeted cells. CPVT is a disease typically caused by a missense mutation in the RYR2 gene that encodes the ryanodine receptor 2 (RYR2), which is found on the sarcoplasmic reticulum (SR) in cardiomyocytes within the heart. This mutation is inherited in an autosomal dominant pattern and leads to incorrect expression of a protein subunit involved in the regulation of calcium release from RYR2, increasing intracellular calcium concentration in the sarcomere 137 . Pan et al. 138 utilized AAV9 for NHEJ-mediated gene disruption in a CPVT mice model with an RYR2 mutation. Mice were treated via subcutaneous injection of AAV9 expressing both SaCas9 and sgRNA targeting exon eight of RYR2 for disruption in cardiomyocytes. CRISPR-Cas9 therapy led to an on-target indel percentage of 9.91%e12.69% with no significant off-target indels occurring. RYR2 disruption led to significant decreases in RYR2 expression and calcium release from SR in cardiomyocytes when compared to control-treated mice. Following adrenergic stimulation, 71% of the control AAV9-treated mice experienced arrhythmia, while arrhythmia was not seen in any of the CRISPR-treated mice. CRISPR-treated mice also showed minimal systemic toxicity. Of the preclinical studies targeting muscle, none included a significant long-term follow up and some did not report off-target indel data. Though the results from each showed encouraging possibilities for CRISPR therapies targeting muscles, the lack of long-term data and missing indel data will require more studies to be completed to validate the long-term efficacy and safety of these therapies. Local injection of CRISPR-Cas9 into the CNS has enabled gene editing for the treatment of multiple devastating neurodegenerative conditions. Both non-viral and viral vectors have been used in treatment of conditions such as Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. Alzheimer's disease is inherited in an autosomal dominant pattern and is characterized by accumulation of amyloid beta plaque and neurofibrillary tangles in the brain, which can lead to synaptic dysfunction, hippocampal neuron loss, and memory impairment 139, 140 . For amyloid beta formation to occur, amyloid precursor protein must first be processed by beta secretase 1 (BACE1), which represents a potential yet difficult therapeutic target 141, 142 . In an effort to ameliorate familial Alzheimer's disease in mice, Park et al. 109 developed a simple but effective amphiphilic peptide nanoparticle to deliver Cas9/sgRNA RNP targeting BACE1 for NHEJ-mediated disruption via intrahippocampal injection. The cationic R7L10 peptide successfully delivered RNP to post-mitotic neuronal cells in mice, with an on-target indel percentage of roughly 45% and without significant off-target indels in nine predicted loci. BACE1 disruption caused a significant reduction in BACE1 expression, percent area of total amyloid beta plaque, and amyloid beta 42 accumulation and secretion. Two intrahippocampal injections showed significantly decreased levels of BACE1 and percent area of amyloid beta when compared to mice treated with a single injection. Treated mice also showed a significant improvement in spatial working memory and associative learning when compared to untreated mice. This therapy demonstrated relative safety, causing minimal increases in inflammation, reactive microglia, blood urea nitrogen, and cleaved caspase 3. Therapeutic and toxic effects of this therapy were measured up until 12 weeks post-injection. Huntington's disease (HD) is caused by a mutation to the huntingtin (HTT ) gene causing expression of an altered HTT protein that is neurotoxic and is commonly inherited in an autosomal dominant pattern 143 . Ekman et al. 144 reported the utility of NHEJ-mediated HTT disruption in treating Huntingtonʹs disease in mice. Mice were given a single intra-striatal injection of AAV1 expressing SaCas9 and sgRNA targeting HTT in medium sized spiny neurons of the brain. This therapy induced an on-target indel percentage of up to 15% with no significant off-target indels occurring at ten predicted loci. HTT disruption led to a significant decrease in HTT protein and mutant HTT proteins, as well as a 15% increase in mean survival when compared to control-treated mice. Additionally, mice with reduced levels of HTT protein showed improved motor function and decreased hind limb clasping. CRISPR-treated mice showed no significant difference in body weight when compared to control-treated mice, as both declined over the course of the 15-week study. Body weight decline is an indicator of HD onset and researchers in this study attributed this outcome in both groups to disease onset prior to AAV-therapy. Genome editing effects were seen for 15 weeks post-injection when all mice eventually died (Fig. 3C) . Amyotrophic lateral sclerosis (ALS) typically causes motor neurons loss and presents with symptoms including muscle atrophy, paralysis, and eventual respiratory dysfunction 4, 145 . ALS has multiple inheritance patterns and its pathogenesis can be attributed to mutations in superoxide dismutase 1 (SOD1), causing a malfunctioning protein which increases the oxidative stress and induces progressive motor neuron loss 146 . Lim et al. 147 have demonstrated the potential of using AAV9 expressing SpCas9 and sgRNA targeting mutant SOD1 to induce nonsense mutations in murine astrocytes of the CNS. Intrathecal injection of the AAV9based CRISPR therapy led to an on-target editing efficiency of about 1.2% in the cervical spinal cord with 4% off-target editing in one of ten predicted loci, though it occurred in a non-protein coding region. SOD1 editing resulted in a significant decrease in SOD1 reactive inclusions in ventral white matter and anterior grey matter when compared to control-treated mice. Additionally, SOD1 editing led to increases in mean survival time and disease duration as well as a higher rotarod percentage, increased motor function, and greater hind limb grip strength when compared to control-treated mice. CRISPR-treated mice showed no signs of discomfort or toxicity throughout the study, indicating the potential safety profile of this therapy. The duration of genome editing effects in this study varied based on survival times which ranged from 128.6 to 150.2 days. All highlighted CNS genome editing studies utilized a local administration technique and showed positive therapeutic outcomes related to each specific disease state. However, these effects were not sustained for more than 6 months. Though these studies have offered promising results, direct local administration enables genome editing in limited areas surrounding the injection sites. Thus, neurological diseases affecting large areas of CNS would require multiple invasive local injections, which increases surgical costs and risk for potential infections. So far, proof-of concept studies illustrating the safety and efficacy of treating neurological diseases by systemic administration of CRISPR-Cas9 system have yet to be reported. CRISPR-Cas9 has found utility for treatment of numerous ocular conditions. Local delivery is desired in this case as a much smaller dose is required. In addition, local injection reduces the potential for off-target editing or adverse events. Delivery with both nonviral and viral delivery systems has showed promise in treating conditions such as X-linked juvenile retinoschisis and age-related macular degeneration. X-linked juvenile retinoschisis is caused by a mutation in retinoschisin 1 (RS1), causing decreased expression by foveal and macular cone photoreceptors as well as bipolar cells 148 . RS1 helps preserve the structure and function of the retina via cell adhesion. Decreased RS1 level can lead to splitting in the macula or surrounding areas of retina, causing reduced signal transduction and progressive vision loss 149 . Chou and colleagues developed a modified polyamidoamine (PAMAM) dendrimer nanoparticle delivery system to encapsulate donor RS1 DNA template plasmids as well as plasmids expressing Cas9/sgRNA targeting RS1 150 . This dendrimer nanoparticle delivery system was administered via single intravitreal injection for RS1 gene insertion via HITI. RS1 gene knock-in was seen at post-injection day 18 and was determined using green fluorescent protein (GFP) signaling. Knock-in persisted until post-injection Day 30 when mice were sacrificed. Analysis of mice eyes showed no histological abnormalities as well as GFP þ ganglion cells in the retina. Ling et al. 71 reported that a single subretinal injection of IDLV vectors successfully enabled genome editing and reduced choroidal neovascularization (CNV) in a murine model of wet age-related macular degeneration. This lentivirus-based therapy delivered SpCas9 mRNA and an sgRNA expression cassette targeting VEGFA in retinal pigment epithelial cells for NHEJmediated disruption. CRISPR-treated mice showed an ontarget indel percentage of up to 44% and no significant off-target indels detected in seven predicted loci. VEGFA disruption led to a significant reduction of both VEGFA expression and CNV area when compared to PBS-treated mice. Seven days following gene editing therapy, mice showed no significant CD3 þ T cell infiltration into retinas or RPE as well as no significant difference in anti-Cas9 IgG when compared to PBS-treated mice. The minimal immunogenicity of this therapy may be due to the route of administration as the eye is considered an immune-privileged organ. Genome editing and therapeutic effects persisted for two weeks post-injection when mice were sacrificed for analysis. For ocular diseases caused by loss-of-function mutations in larger genes, traditional gene replacement therapy or NHEJ-based genome editing therapy cannot use AAV due to its limitation in cargo capacity. Even though dual AAV strategy achieved success in several preclinical studies 44, 55 , the absolute genome editing efficiency in post-mitotic neuronal cells is very low. One of the reasons is that dual AAV strategy does not get every component in target cells at the same time. To improve the genome editing efficiency, Nishiguchi et al. 151 reported a single AAV strategy to enable MMEJ in a murine model of retinal dystrophy. MMEJ utilizes smaller homology arms (about 20 bp) in the packaging construct to allow more room for CRISPR components and donor DNA, to correct vision-impairing mutations 152, 153 . In this study, a single AAV2/8 vector was used to deliver SaCas9, sgRNAs, and donor DNA for mutation replacement. Gnat 1 IRD2/IRD2 Pde6c cpfl1/cpfl1 mice, who are blind due to mutations causing rod and cone cell loss 154 , were treated via subretinal injection with sgRNAs targeting Gnat1 in rod cells for correction of the IRD2 mutation. Successful mutation correction efficiency was between 10% and 11% at 1-and 3-months post-injection, which is much higher than dual AAV strategy. No significant off-target indels were detected at 14 predicted loci. Treated mice showed an increase in rod cell activity as well as improvements in light sensitivity and visual acuity. Though not all preclinical studies demonstrating ocular gene editing provided a long-term follow up, these studies showed positive effects. Utilization of HITI and MMEJ provides an interesting contrast to many other preclinical studies that opted for more traditional editing strategies. In the case of MMEJ, this may be a great option to allow for all-in-one packaging of key CRISPR components to bypass the packaging limit challenges seen with AAV delivery. CRISPR has shown its capability as a genome editing tool in the inner ear. Preclinical studies have demonstrated the feasibility of local delivery into the inner ear using both viral and non-viral vectors. Though inner ear editing is not as widely established as editing in other organs, localized otic CRISPR-Cas9 administration is significant for the treatment of hearing loss disorders to preserve physiological function and improve hearing. Liu's group initially explored the feasibility of CRISPR-Cas9mediated genome editing in the inner ear in a transgenic GFP mouse model 79 . In this proof-of-concept study, cationic lipid transfection reagent Lipofectamine RNAiMAX or Lipofectamine 2000 successfully delivered CRISPR-Cas9 RNP and enabled inner ear genome editing. This paved the way for the following therapeutic studies. In a recent study, the same group demonstrated that cationic liposome-mediated inner ear delivery of CRISPR-Cas9 RNP successfully ameliorated autosomal dominant hearing loss using a Beethoven mouse model where mutant transmembrane channel-like protein 1 (TMC1) causes hearing loss 155 . SpCas9 and sgRNA targeting TMC1 were complexed with Lipofectamine 2000 for delivery via single intracochlear injection. Five days post injection, mice showed an on-target indel percentage ranging from 0.3% to 2.25%, causing successful NHEJ-mediated disruption of TMC1 with no significant off-target indels. TMC1 disruption led to significant preservation of both inner and outer hair cells and significant hearing improvements at eight weeks post-injection. However, mice experienced a decrease in inner hair cell sensory transduction as well as outer hair cell damage following genome editing therapy, possibly due to the well-known cytotoxicity of Lipofectamine 2000. More recently, György et al. 156 showed one time treatment of AAV-mediated CRISPR-Cas9 genome editing prevented autosomal dominant hearing loss in Beethoven mice for up to one year. A single intracochlear injection of AAV2 expressing SaCas9 and sgRNA targeting TMC1 for NHEJ-mediated disruption induced on-target indels ranging from 0.2% (7 days post-injection) to 2.2% (55 days post-injection). Treated mice showed improvements in auditory brainstem response (ABR) for up to 52 weeks postinjection, with one of the treated mice demonstrated an ABR that was indistinguishable from wild type. Twenty-four weeks post injection, treated mice showed preservation of both inner and outer hair cells while untreated mice showed a significant loss in both cell types. This therapy was specific to mutant TMC1 with only 0.0075% indels found at wild type alleles. Though the prevention of hearing loss in this study is encouraging, AAV2-treated mice showed the consequence of reduced inner and outer hair cell mechanosensitivity. These representative studies demonstrated the feasibility, efficacy, and relative safety of inner ear editing with CRISPR, while György et al. 156 took it a step further in showing positive therapeutic effects for a prolonged period of time. Though there are not many preclinical models for genome editing in the inner ear, these studies provide an excellent foundation to build on for future inner ear genome editing therapies. Targeting various cells of the immune system for gene editing has significantly increased the therapeutic versatility of CRISPR-Cas9. Systemic administration of CRISPR for immune modulation is desirable in treating many different conditions such as type two diabetes mellitus and allograft rejection. Wang's group 103 developed a cationic lipid-assisted nanoparticle (CLAN) delivery system for therapeutic genome editing in the immune system of murine models of type II diabetes mellitus (T2DM). CRISPR-Cas9 delivery was accomplished using intravenous injection of CLAN encapsulated Cas9 mRNA and sgRNA, or Cas9/sgRNA encoding plasmids. CLANs were prepared using PEG-PLGA and cationic lipid N,N-bis(2-hydroxyethyl)-N-methyl-N-(2-cholesteryloxycarbonylaminoethyl) ammonium bromide (BHEM-Chol). Treatment of T2DM included CLANs encapsulating SpCas9, sgRNA, and CD68 promoter encoding plasmids that targeted netrin 1 (NTN1) in macrophages and monocytes for NHEJmediated deletion. CRISPR therapy induced 19.6% on-target indels with less than 0.5% indels in five potential off-target loci. NTN1 deletion helped to modulate T2DM-induced inflammatory response by significantly decreasing serum levels of tumor necrosis factor-a (TNF-a), interleukin-6 (IL-6), and adipose tissue levels of netrin-1 and CD11b þ F4/80 þ macrophages. This immune modulation successfully decreased blood glucose and increased insulin sensitivity in T2DM-induced mice with minimal systemic toxicity, however these effects were only reported for 16 days post-treatment. In another study, CLAN encapsulated Cas9 mRNA and sgRNA was tested to ablate cluster of differentiation cell surface receptor 40 (CD40) in dendritic cells (DCs) to induce acute allograft tolerance 100 . NHEJ-mediated disruption of CD40 in mice decreased allograft rejection, prolonged graft survival time, and showed more intact tissue alignment with less graft damage when compared to Rapamycin-treated and untreated mice. It is important to note that not all DCs showed CD40 knockout. Also, all edited DCs eventually were replaced by new ones. The fact that all mice had undergone complete graft rejection after 12 days indicated that there are a number of obstacles to overcome before one can specifically modulate the immune system function by CRSIPR-Cas9-based genome editing. Lastly, CRISPR has shown promise as an adjunct therapy in the treatment of human immunodeficiency virus (HIV) via systemic injection. Dash et al. 157 showed encouraging data related to the use of CRISPR-Cas9 along with long-acting slow-effective release antiretroviral therapy (LASER ART) in treating murine models of HIV. AAV9 expressing SaCas9 and sgRNA targeting HIV1 was given via single intravenous injection for systemic NHEJ-mediated gene deletion. CRISPR-Cas9 therapy induced an on-target indel percentage ranging from 60% to 80% with no detectable off-target indels in 100 predicted loci. Neither HIV1 deletion nor LASER ART therapies alone provided encouraging prospects for long term suppression of HIV1 viral load. When used in combination, CRISPR and LASER ART showed encouraging outcomes as a potential solution for long term viral suppression while further increasing CD4 þ T cells. Combination therapy significantly increased CD4 þ T cells and decreased HIV1 viral load in multiple organs when compared to untreated HIV-induced mice. Additionally, 39% of mice receiving CRISPR/ART combination therapy showed no viral rebound and four of these mice had complete elimination of HIV1 DNA. The study was terminated for specimen analysis at week 14, leaving only five weeks to observe the outcomes following CRISPR-Cas9 therapy. Mancuso et al. 158 furthered the application of CRISPR-Cas9 for in vivo therapeutic genome editing by treating simian immunodeficiency virus (SIV) infected rhesus macaques. Macaques were treated via intravenous injection of AAV9 expressing SaCas9 and sgRNAs targeting the Gag and LTR regions of the SIV viral genome for systemic NHEJ-mediated gene deletion in addition to ART. Treated macaques showed an on-target excision efficiency ranging from 37% to 92% in blood as well as varying levels in other tissues. SIV viral DNA excision led to a 38%e95% decline in proviral DNA found in lymph nodes compared to 20% in a macaque treated with ART alone. Macaques treated with CRISPR and ART did not show any adverse effects following AAV9 inoculation, though no quantification of off-target editing was reported. All genome editing results were determined at 3 weeks following CRISPR therapy when animals were sacrificed for analysis (Fig. 3B) . Eliminating any immunodeficiency virus from hosts is certainly an ambitious attempt, but neither study provided prolonged follow up. Regardless of perceived clinical feasibility, Mancuso et al. 158 showed success in a non-human primate model, which brought in vivo CRISPR-Cas9 therapies one step closer to clinical application. A number of challenges need to be addressed before CRISPR-Cas9 therapeutics can be widely applied in the clinic. These challenges can be classified by several aspects; however, we only discuss delivery-related challenges in this section. Readers are encouraged to read excellent review articles discussing challenges related to the biology 1,159,160 , ethics 161, 162 , and affordability 3, 163 of CRISPR therapeutics. To enable in vivo genome editing using CRISPR-Cas9 system, NHEJ-based gene disruption or deletion requires both Cas9 nucleases and sgRNAs, while HDR-based gene insertion requires Cas9, sgRNA, and exogenous donor DNA, both with proper timing in target cells. Although the knowledge and experience accumulated in protein and gene delivery are extremely helpful, it is clear that in vivo delivery of CRISPR-Cas9 is far more difficult than delivery of proteins and genes alone. For protein and gene delivery, only one therapeutic component, either protein, or DNA/ RNA must be delivered to some target cells. While for CRISPR-Cas9 therapeutics, two or three functional components must present in the same nucleus of a target cell at the same time. This poses additional challenges not only to the delivery efficiency but also to the logistics of the delivery task. 6.1. The in vivo instability of CRISPR-Cas9 therapeutics, which includes biochemistry-related or immunogenicity-related instability The intrinsic biochemical properties of protein and nucleic acid molecules results in poor in vivo stability of Cas9 protein, plasmid DNA, mRNA, sgRNA, as well as exogenous donor DNA. Cas9 protein can be degraded through protease-mediated degradation and hydrolysis in cells and tissues. In addition, when injected into blood, its positive charge can easily recruit negatively charged components of the blood and immune system, leading to fast clearance. The poor in vivo stability of DNA/RNA has been well acknowledged in gene and RNAi delivery 164 . Widespread nucleases in blood and interstitial fluid can quickly degrade DNA/RNA if they are present freely in the body. Therefore, the cargoes must be well protected by delivery vectors. Since exogenous plasmid DNA, RNA, and protein molecules are all immunogenic, when they are administered in vivo, they may elicit host immune responses. The immunogenicity of plasmid DNA can be reduced by removing CpG motifs 165, 166 . In addition, various judicious chemical modifications have decreased the immunogenicity and increased the stability of RNA molecules 167 . The immunogenicity of commonly used Cas9 nucleases poses a different challenge, because they are obtained from Staphylococcus aureus and Streptococcus pyogenes, which are frequently encountered by most humans 21 . Pre-existing anti-Cas9 antibodies have been discovered in humans 22 . One potential solution that may help overcome the challenge is to engineer bacterial Cas9 nucleases by removing their epitopes. Another possible strategy is to look for other suitable CRISPR nucleases. There are enormous efforts on discovering novel CRISPR nucleases, some of them have demonstrated superior genome editing efficiency and accuracy 168 . Nonetheless, the components of the CRISPR-Cas9 system must be fully encapsulated within the delivery vectors to maintain the stability and avoid triggering a host immune response during in vivo transportation. Intravascular AAVs are known to transduce cells in multiple organs and tissues, with limited levels of tissue tropism shown by differing serotypes. Several strategies are being explored to improve the tissue tropism of AAVs, such as direct evolution, discovering novel AAVs from ancient species and creating novel AAVs by engineering capsid proteins 37 . Non-viral vectors greater than several hundred nanometers in diameter are easily blocked by capillary beds in the lungs following intravenous injection, while nanoparticles less than 100 nm are most successful for systemic delivery. Cationic ingredients are frequently used to condense negatively charged DNA and RNA molecules, conferring positive charge to delivery vectors. Following intravenous injection, cationic vectors quickly bind to negatively charged blood components and form large complexes. These complexes are usually blocked in the lungs, leading to rapid clearance from blood circulation and decreased distribution to target tissues. This may be beneficial for pulmonary-targeted therapies, but the positive surface charge must be stably masked when targeting other tissues. Non-viral vectors in blood are also promptly recognized as foreign particles by the host immune system, leading to fast clearance from the bloodstream and decreased distribution to target tissues. PEGylation is a widely used strategy to help shield the surface charge and protect the non-viral vectors from opsonization. It also improves the storage stability of nanoparticles by preventing aggregation. However, the density of PEG chains on nanoparticles should be optimized because too many PEG molecules can inhibit cellular uptake 169e171 . An additional consideration is that PEG itself is immunogenic. The host immune system can produce anti-PEG IgM, which results in accelerated blood clearance of PEGylated vectors during repeated administration 172, 173 . To avoid the immunogenicity issue of PEG, it has been shown that endogenous macromolecules may be used as alternative surface masking molecules 174 . Since DNAs and RNAs are negatively charged hydrophilic macromolecules, they are not able to permeate across the hydrophobic and negatively charged cell membrane. Although viral and non-viral vectors can protect them from degradation in the bloodstream and in tissues, neither shows the desired capability of cell type-specific delivery following intravascular administration. Efforts to achieve cell type-specific gene delivery by non-viral vectors include surface modification by various targeting moieties such as small molecule ligands, aptamers, peptides, and antibodies 175, 176 . For diseases that require systemic administration of CRISPR-Cas9 systems, cell typespecific delivery is ideal to avoid delivery-mediated off-target effects. However, it is worth noting that the clinical success of a genome editing therapy may be achieved for some diseases before a perfect cell type-specific delivery vector is available. This is because repairing the genome of a certain portion of target cells in the desired tissue by local administration of CRISPR-Cas9 therapeutics may bring sufficient clinical benefits to patients while keep the safety risk low. Several preclinical studies highlighted earlier have shown promising therapeutic outcomes after local injection of CRPSPR-Cas9 systems, such as in the inner ear 155 , muscle 118 and brain 109 . Even with these advances, the widespread success of local administration of CRISPR-Cas9 genome editing therapy depends on the disease state, severity, properties of the protein encoded by the target gene, and the desired tissue. Receptor-mediated endocytosis directs CRISPR delivery vectors into early endosomes, which will evolve into late endosomes, and eventually lysosomes, where the low pH environment and digestive enzymes can destroy CRISPR-Cas9 cargoes. Releasing the cargoes into the cytosol before late endosomes/lysosomes are formed is a critical step. It is not a concern for viral vectors, but it is very challenging for non-viral vectors. Different strategies of endosomal escape have been tested, such as cationic lipids-induced instability of the endosome membrane 177 and PEI polymer-induced "proton sponge" effect 178 . However, endosomal release efficiency is still very low 179 . It was reported that membrane fusion peptides and cell penetrating peptides have demonstrated the ability to break endosomal/lysosomal membranes 180, 181 , which may help resolve this issue. Alternatively, direct cytosolic delivery of nanoparticles through membrane fusion bypassed the endosome trap and increased cargo availability in the cytoplasm 174 . For most genome editing applications, the cell nucleus is where CRISPR-Cas9 components perform their function. Dividing cells break up their nuclear membrane during each cell cycle, which allows exogenous large molecules, including Cas9 nucleases, plasmid DNA, mRNA, sgRNA, and donor DNA template, to enter the cell nucleus. However, for non-dividing cells, such as neurons, myocytes, and adipocytes, the nuclear membrane is a formidable barrier. Although it has been demonstrated that nuclear localization sequences could increase the nuclear entry of plasmid DNA 180,182 , more evidence from in vivo studies is needed. Many preclinical and clinical data demonstrated successful genome editing using the CRISPR-Cas9 system in both dividing and nondividing cells. However, the mechanism for Cas9 nucleases to enter cell nuclei is still elusive. Illustrating the sequential process will not only solve a piece of the puzzle regarding CRISPR-Cas9mediated genome editing in mammalian cells, but also shed light on the design of next generation delivery systems. CRISPR-Cas9-mediated genome editing has been recognized as one of the most revolutionary technologies in the 21st century. The precise and permanent repair on the genome of patients' cells will enable a complete cure to numerous inherited and acquired diseases, ranging from cancer, metabolic diseases, nervous system diseases, and infectious diseases. The number of registered clinical trials has been rapidly growing in the past several years (Clinicaltrials.gov). However, the majority of clinical trials are conducted by ex vivo delivery 74 , and only a small number of them use in vivo delivery (Table 2 ). It clearly shows that safe and effective in vivo delivery is the most significant hurdle to widespread clinical success of CRISPR-Cas9 therapeutics. Because the biological nature and mechanism of CRISPR-Cas9 is much more sophisticated than a single gene, RNA, or protein, no existing gene or protein delivery system can be directly adapted for the CRISPR-Cas9 system. Although the experience and lessons learned for protein and gene delivery have been very valuable, CRISPR-Cas9 components need significantly upgraded and innovative in vivo delivery systems. Both viral vectors and non-viral vectors have made remarkable progress for in vivo delivery of CRISPR-Cas9 therapeutics. However, only a small number of studies reported longterm efficacy and benefit of CRISPR-Cas9 genome editing therapies, which should be essential for all genome editing studies. In addition, few non-viral delivery studies reported the loading efficiency of CRISPR-Cas9 cargoes, which makes it difficult to evaluate the potential of the vectors for future development. Nonetheless, appropriate delivery vectors should be designed according to the choice of CRISPR-Cas9 cargoes as discussed earlier, in addition to the route of administration and disease. We are optimistic that different delivery systems will be proven effective for in vivo CRISPR-Cas9-based genome editing therapies. Completely addressing the in vivo delivery challenges requires collaborative efforts from multiple disciplines. Cas9 nuclease engineering might help overcome several challenges, such as reducing its immunogenicity and facilitating endosomal escape as a membrane fusion protein. A Cas9 fusion protein with multiple functions, such as nucleic acids binding, cellular entry, endosomal escaping and nucleus entry, might dramatically change the design of next generation non-viral vectors for CRISPR-Cas9 delivery. Additionally, a clear illustration of biological processes involved in CRISPR-Cas9 genome editing, such as the intranuclear translocation of Cas9 proteins, will help improve the genome editing efficiency. Novel delivery systems which can carry all components of CRISPR-Cas9 in a single vector will most likely win the competition. Cell type-specific systemic delivery remains a challenge in the future and requires novel biomaterials to address the issue. Novel functional lipids, polymers, and peptides as well as viral vectors will continue to enrich the delivery arsenal. In parallel to the development of systemic delivery vectors, local administration should be explored for diseases which have been traditionally considered to be treated by systemic administration. Future efforts should also include efficient delivery of CRISPR-Cas9 to mitochondria for genome editing because many diseases are caused by faulty mitochondrial DNA 183 . The advancement of in vivo CRISPR-Cas9 delivery systems may also dramatically alter the landscape of ex vivo genome editing. Ex vivo genome editing therapy poses a myriad of challenges to cell extraction, culture, manipulation, freezing, and transportation, which are strictly regulated by regulatory authorities. Therefore, it is very possible that when highly specific in vivo delivery technologies are available for clinical genome editing therapy, the need for ex vivo genome editing may dramatically reduce, or even vanish. Special attention must be given to the specificity of in vivo genome editing, which is critical to safe applications of CRISPR-Cas9 therapeutics in the clinic. The consequence of genome modification at off-target sites could be devastating because permanent genome editing at the incorrect sites may deactivate tumor suppressor genes or accidently activate oncogenes, both of which may lead to the development of cancer. In recent years, multiple techniques became available to detect off-target genome editing 184e192 . In the meantime, tremendous efforts have been made to reduce the off-target effects, including engineering Cas9 nucleases such as dimeric Cas9 nucleases 193, 194 , high-fidelity Cas9 195 , enhanced specificity Cas9 196 , hyper-accurate Cas9 197 , Cas9 variants with expanded PAM compatibilities 198 , Cas9 fused with artificial inhibitory domains 199 . Additional efforts were made toward engineering sgRNA molecules, such as truncated sgRNA 200 and paired sgRNAs 201 . As discussed earlier, innovative targeted delivery vectors play vital roles in reducing the delivery-mediated off-target effects. It is important to note that smart non-viral delivery systems may play greater roles even beyond delivery itself, such as reducing off-target genome editing. Stimuli-responsive non-viral delivery systems are promising to enable spatiotemporal genome editing by releasing or activating CRISPR-Cas9 components in target cells only when endogenous or exogenous signals trigger the release 202 . Offtarget genome mutations at unintended sites may be significantly reduced through transient exposure of the cellular genome to CRISPR-Cas9 system. For example, Chen et al. 203 demonstrated that optogenetic regulation of in vivo genome editing minimized the off-target effects by using cationic polymercoated Au nanorods and second near infrared light. It is anticipated that the combination of advanced delivery technologies and molecular engineering technologies will work synergistically to minimize the off-target effects and pave a safe way toward clinical applications. CRISPR-Cas9-mediated genome editing has shown unprecedented power and promise in medicine. Although the lifechanging cures to many diseases seem to be just around the corner, safe and efficient delivery is half the game. Through integration of the knowledge accumulated in delivery science and vector engineering, together with continued collaborative efforts on the development of in vivo delivery systems, CRISPR-Cas9 therapeutics will soon realize their full potential. Therapeutic genome editing: prospects and challenges Genome editing. The new frontier of genome engineering with CRISPR-Cas9 The promise and challenge of therapeutic genome editing The beginning of genomic therapies for ALS Therapeutic developments for Duchenne muscular dystrophy CRISPR-based therapeutic genome editing: strategies and in vivo delivery by AAV vectors CRISPR-based technologies for the manipulation of eukaryotic genomes A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity Unravelling the structural and mechanistic basis of CRISPR-Cas systems CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria RNA-programmed genome editing in human cells Multiplex genome engineering using CRISPR/Cas systems DNA interrogation by the CRISPR RNA-guided endonuclease Cas9 Conformational control of DNA target cleavage by CRISPR-Cas9 The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway MMEJ repair of double-strand breaks (directors cut): deleted sequences and alternative endings MMEJassisted gene knock-in using TALENs and CRISPR-Cas9 with the PITCh systems Homologymediated end joining-based targeted integration using CRISPR/Cas9 In vivo genome editing via CRISPR/Cas9 mediated homologyindependent targeted integration Cas9 immunity creates challenges for CRISPR gene editing therapies Identification of preexisting adaptive immunity to Cas9 proteins in humans Optimization of CRISPR/Cas9 delivery to human hematopoietic stem and progenitor cells for therapeutic genomic rearrangements Rapid and tunable method to temporally control gene editing based on conditional Cas9 stabilization Targeted gene knock-in by homology-directed genome editing using Cas9 ribonucleoprotein and AAV donor delivery CRISPR-READI: efficient generation of knockin mice by CRISPR RNP electroporation and AAV donor infection CRISPR-mediated integration of large gene cassettes using AAV donor vectors Virus-mediated genome editing via homology-directed repair in mitotic and postmitotic cells in mammalian brain CRISPR/Cas9-based genome editing for disease modeling and therapy: challenges and opportunities for nonviral delivery Engineering the delivery system for CRISPR-based genome editing Delivery of genome-editing biomacromolecules for treatment of lung genetic disorders Delivery technologies for genome editing Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics Gene therapy Entering the modern era of gene therapy Turning genes into medicinesdwhat have we learned from gene therapy drug development in the past decade? Adeno-associated virus vector as a platform for gene therapy delivery Adeno-associated virus at 50: a golden anniversary of discovery, research, and gene therapy successda personal perspective Birth of a new therapeutic platform: 47 years of adenoassociated virus biology from virus discovery to licensed gene therapy Adeno-associated virus biology Engineering adeno-associated virus vectors for gene therapy In vivo genome editing using Staphylococcus aureus Cas9 Characterization of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice AAV-mediated CRISPR/Cas gene editing of retinal cells in vivo Evaluation and reduction of CRISPR offtarget cleavage events Adenovirus-adenoassociated virus hybrid for large-scale recombinant adeno-associated virus production Adeno-associated virus vectorology, manufacturing, and clinical applications Scalable downstream strategies for purification of recombinant adeno-associated virus vectors in light of the properties Curing hemophilia A by NHEJ-mediated ectopic F8 insertion in the mouse Hemophilia A ameliorated in mice by CRISPR-based in vivo genome editing of human Factor VIII CRISPR-Cas9-mediated in vivo gene integration at the albumin locus recovers hemostasis in neonatal and adult hemophilia B mice AAV-mediated CRISPR/Cas9 gene editing in murine phenylketonuria Treatment of a metabolic liver disease by in vivo genome base editing in adult mice A mutationindependent CRISPR-Cas9-mediated gene targeting approach to treat a murine model of ornithine transcarbamylase deficiency Enhanced CRISPR-Cas9 correction of Duchenne muscular dystrophy in mice by a self-complementary AAV delivery system Life-Long AAV-mediated CRISPR genome editing in dystrophic heart improves cardiomyopathy without causing serious lesions in MDX mice Functional rescue of dystrophin deficiency in mice caused by frameshift mutations using Campylobacter jejuni Cas9 In vivo CRISPR/Cas9-Mediated genome editing mitigates photoreceptor degeneration in a mouse model of X-linked retinitis pigmentosa In situ gene therapy via AAV-CRISPR-Cas9-mediated targeted gene regulation Productive human immunodeficiency virus type 1 (HIV-1) infection of nonproliferating human monocytes Lentiviruses as gene transfer agents for delivery to nondividing cells In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector Transduction of human CD34 þ cells that mediate long-term engraftment of NOD/SCID mice by HIV vectors A third-generation lentivirus vector with a conditional packaging system Lentiviral vectors, two decades later In vivo gene therapy of metachromatic leukodystrophy by lentiviral vectors: correction of neuropathology and protection against learning impairments in affected mice Stable knockdown of microRNA in vivo by lentiviral vectors State-of-the-art gene-based therapies: the road ahead Ex vivo gene transfer and correction for cell-based therapies Lentiviral delivery of co-packaged Cas9 mRNA and a VEGFa-targeting guide RNA prevents wet age-related macular degeneration in mice Targeting mutant KRAS with CRISPR-Cas9 controls tumor growth ABL genomic editing sufficiently abolishes oncogenesis of human chronic myeloid leukemia cells in vitro and in vivo Delivery of tissue-targeted scalpels: opportunities and challenges for in vivo CRISPR/Cas-based genome editing Drug delivery systems: entering the mainstream The mechanism of vesicle formation Lipid nanoparticle technology for therapeutic gene regulation in the liver Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo Global CNS gene transfer for a childhood neurogenetic enzyme deficiency: canavan disease Aspartoacylase gene transfer to the mammalian central nervous system with therapeutic implications for Canavan disease Pfizer-BioNTech covid-19 vaccine FDA EAU letter of authorization US Food and Drug Administration. Moderna COVID-19 vaccine EUA letter of authorization CRISPR-Cas9 genome editing using targeted lipid nanoparticles for cancer therapy Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves liver-specific in vivo genome editing of Angptl3 Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA Fast and efficient CRISPR/Cas9 genome editing in vivo enabled by bioreducible lipid and messenger RNA nanoparticles Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles A Single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing Lipid-based nanoparticle technologies for liver targeting Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing Nanoparticles in the clinic: an update A biodegradable nanocapsule delivers a Cas9 ribonucleoprotein complex for in vivo genome editing Genome editing of mutant KRAS through supramolecular polymer-mediated delivery of Cas9 ribonucleoprotein for colorectal cancer therapy Artificial virus delivers CRISPR-Cas9 system for genome editing of cells in mice Therapeutic genome editing of triple-negative breast tumors using a noncationic and deformable nanolipogel Cdk 5 knocking out mediated by CRISPR-Cas9 genome editing for PD-L1 attenuation and enhanced antitumor immunity Targeting of NLRP3 inflammasome with gene editing for the amelioration of inflammatory diseases In situ repurposing of dendritic cells with CRISPR/Cas9-based nanomedicine to induce transplant tolerance Carboxylated branched poly(beta-amino ester) nanoparticles enable robust cytosolic protein delivery and CRISPR-Cas9 gene editing Optimization of lipid-assisted nanoparticle for disturbing neutrophils-related inflammation Macrophagespecific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications Peptide self-assembled nanostructures for drug delivery applications Endosome-disruptive peptides for improving cytosolic delivery of bioactive macromolecules Engineered amphiphilic peptides enable delivery of proteins and CRISPR-associated nucleases to airway epithelia CRISPR-delivery particles targeting nuclear receptorinteracting protein 1 (NRIP1) in adipose cells to enhance energy expenditure In vivo neuronal gene editing via CRISPR-Cas9 amphiphilic nanocomplexes alleviates deficits in mouse models of Alzheimer's disease Peptide/Cas9 nanostructures for ribonucleoprotein cell membrane transport and gene edition Nonviral gene editing via CRISPR/Cas9 delivery by membrane-disruptive and endosomolytic helical polypeptide Enzymatic noncovalent synthesis for mitochondrial genetic engineering of cancer cells Gold nanoparticles for nucleic acid delivery Gold nanoparticles for biology and medicine Immunological properties of gold nanoparticles Targeting dendritic cells through gold nanoparticles: a review on the cellular uptake and subsequent immunological properties Genome editing for cancer therapy: delivery of Cas9 protein/sgRNA Plasmid via a gold nanocluster Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair Mesoporous silica nanoparticles for drug and gene delivery A versatile nonviral delivery system for multiplex gene-editing in the liver Co-delivery of sorafenib and CRISPR/Cas9 based on targeted core-shell hollow mesoporous organosilica nanoparticles for synergistic HCC therapy Near-infrared upconversion-activated CRISPR-Cas9 system: a remote-controlled gene editing platform Virus-like nanoparticle as a co-delivery system to enhance efficacy of CRISPR/Cas9-based cancer immunotherapy A pH-responsive silica-metal-organic framework hybrid nanoparticle for the delivery of hydrophilic drugs, nucleic acids, and CRISPR-Cas9 genome-editing machineries Engineering of monosized lipid-coated mesoporous silica nanoparticles for CRISPR delivery Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype Mutations in PCSK9 cause autosomal dominant hypercholesterolemia The clinical, biochemical, and molecular spectrum of ornithine transcarbamylase deficiency The biochemical and molecular spectrum of ornithine transcarbamylase deficiency Codelivery of CRISPR-Cas9 and chlorin e6 for spatially controlled tumor-specific gene editing with synergistic drug effects Cdk 5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity Cancer immunotherapy comes of age and looks for maturity Delivery technologies for cancer immunotherapy The dystrophin complex: structure, function, and implications for therapy Extracellular nanovesicles for packaging of CRISPR-Cas9 protein and sgRNA to induce therapeutic exon skipping Intracellular calcium release and cardiac disease In vivo Ryr2 editing corrects catecholaminergic polymorphic ventricular tachycardia A systemic view of Alzheimer diseasedinsights from amyloid-beta metabolism beyond the brain Cerebral amyloid angiopathy and Alzheimer diseasedone peptide, two pathways Targeting the beta secretase BACE1 for Alzheimer's disease therapy BACE1: the beta-secretase enzyme in Alzheimer's disease Huntington disease CRISPR-Cas9-mediated genome editing increases lifespan and improves motor deficits in a Huntington's disease mouse model Amyotrophic lateral sclerosis Amyotrophic lateral sclerosis Treatment of a mouse model of ALS by in vivo base editing X-linked juvenile retinoschisis: clinical diagnosis, genetic analysis, and molecular mechanisms X-linked juvenile retinoschisis: phenotypic and genetic characterization Dual supramolecular nanoparticle vectors enable CRISPR/Cas9-mediated knockin of retinoschisin 1 geneda potential nonviral therapeutic solution for X-Linked juvenile retinoschisis Single AAV-mediated mutation replacement genome editing in limited number of photoreceptors restores vision in mice Mechanism of microhomology-mediated end-joining promoted by human DNA polymerase theta Microhomology-mediated end joining and homologous recombination share the initial end resection step to repair DNA double-strand breaks in mammalian cells Evolution of phototransduction, vertebrate photoreceptors and retina Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents Allele-specific gene editing prevents deafness in a model of dominant progressive hearing loss Sequential LASER ART and CRISPR treatments eliminate HIV-1 in a subset of infected humanized mice CRISPR based editing of SIV proviral DNA in ART treated non-human primates CRISPR-Cas guides the future of genetic engineering CRISPR-based technologies for the manipulation of eukaryotic genomes No time to wastedthe ethical challenges created by CRISPR: CRISPR/Cas, being an efficient, simple, and cheap technology to edit the genome of any organism, raises many ethical and regulatory issues beyond the use to manipulate human germ line cells CRISPR ethics: moral considerations for applications of a powerful tool The daunting economics of therapeutic genome editing Progress in systemic co-delivery of microRNAs and chemotherapeutics for cancer treatment by using lipid-based nanoparticles Contribution of CpG motifs to the immunogenicity of DNA vaccines Plasmid CpG depletion improves degree and duration of tumor gene expression after intravenous administration of polyplexes Structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing Discovery and functional characterization of diverse class 2 CRISPR-Cas systems Effect of pegylation on pharmaceuticals Three significant highlights of controlled drug delivery over the past 55 years: PEGylation, ADCs, and EPR PEGylation as a strategy for improving nanoparticle-based drug and gene delivery Accelerated blood clearance of PEGylated liposomes following preceding liposome injection: effects of lipid dose and PEG surfacedensity and chain length of the first-dose liposomes Influence of the physicochemical properties of liposomes on the accelerated blood clearance phenomenon in rats Cytosolic codelivery of miRNA-34a and docetaxel with core-shell nanocarriers via caveolae-mediated pathway for the treatment of metastatic breast cancer Targeting strategies for tissue-specific drug delivery Engineering precision nanoparticles for drug delivery Mechanism of DNA release from cationic liposome/DNA complexes used in cell transfection The Proton sponge: a trick to enter cells the viruses did not exploit A window onto siRNA delivery Multifunctional peptide-PEG intercalating conjugates: programmatic of gene delivery to the bloodebrain barrier GALA: a designed synthetic pHresponsive amphipathic peptide with applications in drug and gene delivery Short biodegradable polyamines for gene delivery and transfection of brain capillary endothelial cells The special considerations of gene therapy for mitochondrial diseases Off-target effects in CRISPR/Cas9-mediated genome engineering Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq CRISPR offtarget detection with DISCOVER-seq Defining CRISPR-Cas9 genome-wide nuclease activities with CIRCLE-seq Profiling Genome-wide specificity of CRISPR-Cas9 using digenome-seq Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases In vivo CRISPR editing with no detectable genomewide off-target mutations Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects Rationally engineered Cas9 nucleases with improved specificity Enhanced proofreading governs CRISPR-Cas9 targeting accuracy Evolved Cas9 variants with broad PAM compatibility and high DNA specificity Coupling Cas9 to artificial inhibitory domains enhances CRISPR-Cas9 target specificity Improving CRISPR-Cas nuclease specificity using truncated guide RNAs Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity Spatiotemporal delivery of CRISPR/Cas9 genome editing machinery using stimuli-responsive vehicles Near-infrared optogenetic engineering of photothermal nanoCRISPR for programmable genome editing