Research Progress on Extracellular Vesicle Delivery CRISPR/Cas9 System

Kun LIU, Yanqing LIU, Nan HUANG, Xi YANG, Qiaoli LANG, Liangpeng GE, Rendong FANG

China Biotechnology ›› 2024, Vol. 44 ›› Issue (6) : 1-10.

PDF(593 KB)
PDF(593 KB)
China Biotechnology ›› 2024, Vol. 44 ›› Issue (6) : 1-10. DOI: 10.13523/j.cb.2401053

Research Progress on Extracellular Vesicle Delivery CRISPR/Cas9 System

Author information +
History +

Abstract

The CRISPR/Cas9 system, known for its efficient and easy gene-editing capabilities, is widely preferred and extensively used in various fields, attracting considerable attention from researchers both domestically and internationally. Extracellular vesicles, as natural nanoscale carriers with abundant sources, are proving to be a highly attractive delivery vehicle for the CRISPR/Cas9 system. Compared to conventional viral or non-viral vectors, extracellular vesicles offer significant advantages in terms of safety, capacity, penetrability, targeting specificity, and transformative potential. They are poised to become the optimal carriers for delivery of the CRISPR/Cas9 system. This article summarizes common delivery strategies and loading methods for the CRISPR/Cas9 system, contrasts extracellular vesicles with other carriers, and provides a comprehensive review of the advantages, domestic and international research progress, and applications of extracellular vesicles in delivering the CRISPR/Cas9 system. The goal is to contribute to the advancement of the field of gene-editing delivery.

Key words

CRISPR/Cas9 system / Geneme editing / Extracellular vesicles / Nanocarriers

Cite this article

Download Citations
Kun LIU, Yanqing LIU, Nan HUANG, et al. Research Progress on Extracellular Vesicle Delivery CRISPR/Cas9 System[J]. China Biotechnology, 2024, 44(6): 1-10 https://doi.org/10.13523/j.cb.2401053

References

[1]
Kalluri R, LeBleu V S. The biology, function, and biomedical applications of exosomes. Science, 2020, 367(6478): eaau6977.
[2]
Ishino Y, Shinagawa H, Makino K, et al. Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. Journal of Bacteriology, 1987, 169(12): 5429-5433.
The iap gene in Escherichia coli is responsible for the isozyme conversion of alkaline phosphatase. We analyzed the 1,664-nucleotide sequence of a chromosomal DNA segment that contained the iap gene and its flanking regions. The predicted iap product contained 345 amino acids with an estimated molecular weight of 37,919. The 24-amino-acid sequence at the amino terminus showed features characteristic of a signal peptide. Two proteins of different sizes were identified by the maxicell method, one corresponding to the Iap protein and the other corresponding to the processed product without the signal peptide. Neither the isozyme-converting activity nor labeled Iap proteins were detected in the osmotic-shock fluid of cells carrying a multicopy iap plasmid. The Iap protein seems to be associated with the membrane.
[3]
Jansen R, van Embden J D A, Gaastra W, et al. Identification of genes that are associated with DNA repeats in prokaryotes. Molecular Microbiology, 2002, 43(6): 1565-1575.
Using in silico analysis we studied a novel family of repetitive DNA sequences that is present among both domains of the prokaryotes (Archaea and Bacteria), but absent from eukaryotes or viruses. This family is characterized by direct repeats, varying in size from 21 to 37 bp, interspaced by similarly sized non-repetitive sequences. To appreciate their characteri-stic structure, we will refer to this family as the clustered regularly interspaced short palindromic repeats (CRISPR). In most species with two or more CRISPR loci, these loci were flanked on one side by a common leader sequence of 300-500 b. The direct repeats and the leader sequences were conserved within a species, but dissimilar between species. The presence of multiple chromosomal CRISPR loci suggests that CRISPRs are mobile elements. Four CRISPR-associated (cas) genes were identified in CRISPR-containing prokaryotes that were absent from CRISPR-negative prokaryotes. The cas genes were invariably located adjacent to a CRISPR locus, indicating that the cas genes and CRISPR loci have a functional relationship. The cas3 gene showed motifs characteristic for helicases of the superfamily 2, and the cas4 gene showed motifs of the RecB family of exonucleases, suggesting that these genes are involved in DNA metabolism or gene expression. The spatial coherence of CRISPR and cas genes may stimulate new research on the genesis and biological role of these repeats and genes.
[4]
Doudna J A, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science, 2014, 346(6213): e1258096.
[5]
Sternberg S H, Haurwitz R E, Doudna J A. Mechanism of substrate selection by a highly specific CRISPR endoribonuclease. RNA, 2012, 18(4): 661-672.
Bacteria and archaea possess adaptive immune systems that rely on small RNAs for defense against invasive genetic elements. CRISPR (clustered regularly interspaced short palindromic repeats) genomic loci are transcribed as long precursor RNAs, which must be enzymatically cleaved to generate mature CRISPR-derived RNAs (crRNAs) that serve as guides for foreign nucleic acid targeting and degradation. This processing occurs within the repetitive sequence and is catalyzed by a dedicated Cas6 family member in many CRISPR systems. In Pseudomonas aeruginosa, crRNA biogenesis requires the endoribonuclease Csy4 (Cas6f), which binds and cleaves at the 3' side of a stable RNA stem-loop structure encoded by the CRISPR repeat. We show here that Csy4 recognizes its RNA substrate with an ~50 pM equilibrium dissociation constant, making it one of the highest-affinity protein:RNA interactions of this size reported to date. Tight binding is mediated exclusively by interactions upstream of the scissile phosphate that allow Csy4 to remain bound to its product and thereby sequester the crRNA for downstream targeting. Substrate specificity is achieved by RNA major groove contacts that are highly sensitive to helical geometry, as well as a strict preference for guanosine adjacent to the scissile phosphate in the active site. Collectively, our data highlight diverse modes of substrate recognition employed by Csy4 to enable accurate selection of CRISPR transcripts while avoiding spurious, off-target RNA binding and cleavage.
[6]
Cong L, Ran F A, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science, 2013, 339(6121): 819-823.
Functional elucidation of causal genetic variants and elements requires precise genome editing technologies. The type II prokaryotic CRISPR (clustered regularly interspaced short palindromic repeats)/Cas adaptive immune system has been shown to facilitate RNA-guided site-specific DNA cleavage. We engineered two different type II CRISPR/Cas systems and demonstrate that Cas9 nucleases can be directed by short RNAs to induce precise cleavage at endogenous genomic loci in human and mouse cells. Cas9 can also be converted into a nicking enzyme to facilitate homology-directed repair with minimal mutagenic activity. Lastly, multiple guide sequences can be encoded into a single CRISPR array to enable simultaneous editing of several sites within the mammalian genome, demonstrating easy programmability and wide applicability of the RNA-guided nuclease technology.
[7]
Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science, 2012, 337(6096): 816-821.
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems provide bacteria and archaea with adaptive immunity against viruses and plasmids by using CRISPR RNAs (crRNAs) to guide the silencing of invading nucleic acids. We show here that in a subset of these systems, the mature crRNA that is base-paired to trans-activating crRNA (tracrRNA) forms a two-RNA structure that directs the CRISPR-associated protein Cas9 to introduce double-stranded (ds) breaks in target DNA. At sites complementary to the crRNA-guide sequence, the Cas9 HNH nuclease domain cleaves the complementary strand, whereas the Cas9 RuvC-like domain cleaves the noncomplementary strand. The dual-tracrRNA:crRNA, when engineered as a single RNA chimera, also directs sequence-specific Cas9 dsDNA cleavage. Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNA-programmable genome editing.
[8]
Sternberg S H, Redding S, Jinek M, et al. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature, 2014, 507: 62-67.
[9]
Zhang B H. CRISPR/Cas gene therapy. Journal of Cellular Physiology, 2021, 236(4): 2459-2481.
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated enzyme (Cas) is a naturally occurring genome editing tool adopted from the prokaryotic adaptive immune defense system. Currently, CRISPR/Cas9-based genome editing has been becoming one of the most promising tools for treating human genetic diseases, including cardiovascular diseases, neuro-disorders, and cancers. As the quick modification of the CRISPR/Cas9 system, including delivery system, CRISPR/Cas9-based gene therapy has been extensively studied in preclinic and clinic treatments. CRISPR/Cas genome editing is also a robust tool to create animal genetic models for studying and treating human genetic disorders, particularly diseases associated with point mutations. However, significant challenges also remain before CRISPR/Cas technology can be routinely employed in the clinic for treating different genetic diseases, which include toxicity and immune response of treated cells to CRISPR/Cas component, highly throughput delivery method, and potential off-target impact. The off-target effect is one of the major concerns for CRISPR/Cas9 gene therapy, more research should be focused on limiting this impact by designing high specific gRNAs and using high specificity of Cas enzymes. Modifying the CRISPR/Cas9 delivery method not only targets a specific tissue/cell but also potentially limits the off-target impact.© 2020 Wiley Periodicals LLC.
[10]
Lieber M R. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annual Review of Biochemistry, 2010, 79: 181-211.
Double-strand DNA breaks are common events in eukaryotic cells, and there are two major pathways for repairing them: homologous recombination (HR) and nonhomologous DNA end joining (NHEJ). The various causes of double-strand breaks (DSBs) result in a diverse chemistry of DNA ends that must be repaired. Across NHEJ evolution, the enzymes of the NHEJ pathway exhibit a remarkable degree of structural tolerance in the range of DNA end substrate configurations upon which they can act. In vertebrate cells, the nuclease, DNA polymerases, and ligase of NHEJ are the most mechanistically flexible and multifunctional enzymes in each of their classes. Unlike repair pathways for more defined lesions, NHEJ repair enzymes act iteratively, act in any order, and can function independently of one another at each of the two DNA ends being joined. NHEJ is critical not only for the repair of pathologic DSBs as in chromosomal translocations, but also for the repair of physiologic DSBs created during variable (diversity) joining [V(D)J] recombination and class switch recombination (CSR). Therefore, patients lacking normal NHEJ are not only sensitive to ionizing radiation (IR), but also severely immunodeficient.
[11]
Liu S, Wu X, Chandra S, et al. Extracellular vesicles: emerging tools as therapeutic agent carriers. Acta Pharmaceutica Sinica B, 2022, 12(10): 3822-3842.
Extracellular vesicles (EVs) are secreted by both eukaryotes and prokaryotes, and are present in all biological fluids of vertebrates, where they transfer DNA, RNA, proteins, lipids, and metabolites from donor to recipient cells in cell-to-cell communication. Some EV components can also indicate the type and biological status of their parent cells and serve as diagnostic targets for liquid biopsy. EVs can also natively carry or be modified to contain therapeutic agents (, nucleic acids, proteins, polysaccharides, and small molecules) by physical, chemical, or bioengineering strategies. Due to their excellent biocompatibility and stability, EVs are ideal nanocarriers for bioactive ingredients to induce signal transduction, immunoregulation, or other therapeutic effects, which can be targeted to specific cell types. Herein, we review EV classification, intercellular communication, isolation, and characterization strategies as they apply to EV therapeutics. This review focuses on recent advances in EV applications as therapeutic carriers from research towards animal models and early clinical applications, using representative examples in the fields of cancer chemotherapeutic drug, cancer vaccine, infectious disease vaccines, regenerative medicine and gene therapy. Finally, we discuss current challenges for EV therapeutics and their future development.© 2022 Chinese Pharmaceutical Association and Institute of Materia Medica, Chinese Academy of Medical Sciences. Production and hosting by Elsevier B.V.
[12]
He C J, Zheng S, Luo Y, et al. Exosome theranostics: biology and translational medicine. Theranostics, 2018, 8(1): 237-255.
Exosomes are common membrane-bound nanovesicles that contain diverse biomolecules, such as lipids, proteins, and nucleic acids. Exosomes are derived from cells through exocytosis, are ingested by target cells, and can transfer biological signals between local or distant cells. Exosome secretion is a constitutive phenomenon that is involved in both physiological and pathological processes and determines both the exosomal surface molecules and the contents. Hence, we can exploit exosomes as biomarkers, vaccines and drug carriers and modify them rationally for therapeutic interventions. However, it is still a challenge to identify, isolate and quantify exosomes accurately, efficiently and selectively. Further studies on exosomes will explore their potential in translational medicine and provide new avenues for the creation of effective clinical diagnostics and therapeutic strategies; the use of exosomes in these applications can be called exosome theranostics. This review describes the fundamental processes of exosome formation and uptake. In addition, the physiological and pathological roles of exosomes in biology are also illustrated with a focus on how exosomes can be exploited or engineered as powerful tools in translational medicine.
[13]
吴忧, 辛林. 新的药物传递系统: 外泌体作为药物载体递送. 中国生物工程杂志, 2020, 40(9): 28-35.
Wu Y, Xin L. New drug delivery system: delivery of exosomes as drug carriers. China Biotechnology, 2020, 40(9): 28-35.
[14]
Pofali P, Mondal A, Londhe V. Exosome as a natural gene delivery vector for cancer treatment. Current Cancer Drug Targets, 2020, 20(11): 821-830.
[15]
Montagna C, Petris G, Casini A, et al. VSV-G-enveloped vesicles for traceless delivery of CRISPR-Cas9. Molecular Therapy Nucleic Acids, 2018, 12: 453-462.
[16]
Yip B H. Recent advances in CRISPR/Cas9 delivery strategies. Biomolecules, 2020, 10(6): 839.
[17]
Horii T, Arai Y, Yamazaki M, et al. Validation of microinjection methods for generating knockout mice by CRISPR/Cas-mediated genome engineering. Scientific Reports, 2014, 4: 4513.
The CRISPR/Cas system, in which the Cas9 endonuclease and a guide RNA complementary to the target are sufficient for RNA-guided cleavage of the target DNA, is a powerful new approach recently developed for targeted gene disruption in various animal models. However, there is little verification of microinjection methods for generating knockout mice using this approach. Here, we report the verification of microinjection methods of the CRISPR/Cas system. We compared three methods for injection: (1) injection of DNA into the pronucleus, (2) injection of RNA into the pronucleus, and (3) injection of RNA into the cytoplasm. We found that injection of RNA into the cytoplasm was the most efficient method in terms of the numbers of viable blastocyst stage embryos and full-term pups generated. This method also showed the best overall knockout efficiency.
[18]
Long C Z, McAnally J R, Shelton J M, et al. Prevention of muscular dystrophy in mice by CRISPR/Cas9-mediated editing of germline DNA. Science, 2014, 345(6201): 1184-1188.
Duchenne muscular dystrophy (DMD) is an inherited X-linked disease caused by mutations in the gene encoding dystrophin, a protein required for muscle fiber integrity. DMD is characterized by progressive muscle weakness and a shortened life span, and there is no effective treatment. We used clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9)-mediated genome editing to correct the dystrophin gene (Dmd) mutation in the germ line of mdx mice, a model for DMD, and then monitored muscle structure and function. Genome editing produced genetically mosaic animals containing 2 to 100% correction of the Dmd gene. The degree of muscle phenotypic rescue in mosaic mice exceeded the efficiency of gene correction, likely reflecting an advantage of the corrected cells and their contribution to regenerating muscle. With the anticipated technological advances that will facilitate genome editing of postnatal somatic cells, this strategy may one day allow correction of disease-causing mutations in the muscle tissue of patients with DMD. Copyright © 2014, American Association for the Advancement of Science.
[19]
Liu C, Zhang L, Liu H, et al. Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications. Journal of Controlled Release, 2017, 266: 17-26.
The CRISPR-Cas9 genome-editing system is a part of the adaptive immune system in archaea and bacteria to defend against invasive nucleic acids from phages and plasmids. The single guide RNA (sgRNA) of the system recognizes its target sequence in the genome, and the Cas9 nuclease of the system acts as a pair of scissors to cleave the double strands of DNA. Since its discovery, CRISPR-Cas9 has become the most robust platform for genome engineering in eukaryotic cells. Recently, the CRISPR-Cas9 system has triggered enormous interest in therapeutic applications. CRISPR-Cas9 can be applied to correct disease-causing gene mutations or engineer T cells for cancer immunotherapy. The first clinical trial using the CRISPR-Cas9 technology was conducted in 2016. Despite the great promise of the CRISPR-Cas9 technology, several challenges remain to be tackled before its successful applications for human patients. The greatest challenge is the safe and efficient delivery of the CRISPR-Cas9 genome-editing system to target cells in human body. In this review, we will introduce the molecular mechanism and different strategies to edit genes using the CRISPR-Cas9 system. We will then highlight the current systems that have been developed to deliver CRISPR-Cas9 in vitro and in vivo for various therapeutic purposes.Copyright © 2017 Elsevier B.V. All rights reserved.
[20]
Hayashi H, Kubo Y, Izumida M, et al. Efficient viral delivery of Cas 9 into human safe harbor. Scientific Reports, 2020, 10: 21474.
Gene editing using CRISPR/Cas9 is a promising method to cure many human genetic diseases. We have developed an efficient system to deliver Cas9 into the adeno-associated virus integration site 1 (AAVS1) locus, known as a safe harbor, using lentivirus and AAV viral vectors, as a step toward future in vivo transduction. First, we introduced Cas9v1 (derived from Streptococcus pyogenes) at random into the genome using a lentiviral vector. Cas9v1 activity was used when the N-terminal 1.9 kb, and C-terminal 2.3 kb fragments of another Cas9v2 (human codon-optimized) were employed sequentially with specific single-guide RNAs (sgRNAs) and homology donors carried by AAV vectors into the AAVS1 locus. Then, Cas9v1 was removed from the genome by another AAV vector containing sgRNA targeting the long terminal repeat of the lentivirus vector. The reconstituted Cas9v2 in the AAVS1 locus was functional and gene editing was efficient.
[21]
Fang H Q, Bygrave A M, Roth R H, et al. An optimized CRISPR/Cas 9 approach for precise genome editing in neurons. eLife, 2021, 10: e65202.
[22]
Anzalone A V, Koblan L W, Liu D R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nature Biotechnology, 2020, 38: 824-844.
The development of new CRISPR-Cas genome editing tools continues to drive major advances in the life sciences. Four classes of CRISPR-Cas-derived genome editing agents-nucleases, base editors, transposases/recombinases and prime editors-are currently available for modifying genomes in experimental systems. Some of these agents have also moved rapidly into the clinic. Each tool comes with its own capabilities and limitations, and major efforts have broadened their editing capabilities, expanded their targeting scope and improved editing specificity. We analyze key considerations when choosing genome editing agents and identify opportunities for future improvements and applications in basic research and therapeutics.
[23]
Liu P P, Liang S Q, Zheng C W, et al. Improved prime editors enable pathogenic allele correction and cancer modelling in adult mice. Nature Communications, 2021, 12: 2121.
Prime editors (PEs) mediate genome modification without utilizing double-stranded DNA breaks or exogenous donor DNA as a template. PEs facilitate nucleotide substitutions or local insertions or deletions within the genome based on the template sequence encoded within the prime editing guide RNA (pegRNA). However, the efficacy of prime editing in adult mice has not been established. Here we report an NLS-optimized SpCas9-based prime editor that improves genome editing efficiency in both fluorescent reporter cells and at endogenous loci in cultured cell lines. Using this genome modification system, we could also seed tumor formation through somatic cell editing in the adult mouse. Finally, we successfully utilize dual adeno-associated virus (AAVs) for the delivery of a split-intein prime editor and demonstrate that this system enables the correction of a pathogenic mutation in the mouse liver. Our findings further establish the broad potential of this genome editing technology for the directed installation of sequence modifications in vivo, with important implications for disease modeling and correction.
[24]
Rok M, Wong T W Y, Maino E, et al. Prevention of early-onset cardiomyopathy in Dmd exon 52-54 deletion mice by CRISPR-Cas9-mediated exon skipping. Molecular Therapy Methods & Clinical Development, 2023, 30: 246-258.
[25]
Srivastava A, Mallela K M G, Deorkar N, et al. Manufacturing challenges and rational formulation development for AAV viral vectors. Journal of Pharmaceutical Sciences, 2021, 110(7): 2609-2624.
Adeno-associated virus (AAV) has emerged as a leading platform for gene delivery for treating various diseases due to its excellent safety profile and efficient transduction to various target tissues. However, the large-scale production and long-term storage of viral vectors is not efficient resulting in lower yields, moderate purity, and shorter shelf-life compared to recombinant protein therapeutics. This review provides a comprehensive analysis of upstream, downstream and formulation unit operation challenges encountered during AAV vector manufacturing, and discusses how desired product quality attributes can be maintained throughout product shelf-life by understanding the degradation mechanisms and formulation strategies. The mechanisms of various physical and chemical instabilities that the viral vector may encounter during its production and shelf-life because of various stressed conditions such as thermal, shear, freeze-thaw, and light exposure are highlighted. The role of buffer, pH, excipients, and impurities on the stability of viral vectors are also discussed. As such, the aim of this review is to outline the tools and a potential roadmap for improving the quality of AAV-based drug products by stressing the need for a mechanistic understanding of the involved processes.Copyright © 2021. Published by Elsevier Inc.
[26]
Kantor A, McClements M E, MacLaren R E. CRISPR-Cas9 DNA base-editing and prime-editing. International Journal of Molecular Sciences, 2020, 21(17): 6240.
[27]
Ahmadi S E, Soleymani M, Shahriyary F, et al. Viral vectors and extracellular vesicles: innate delivery systems utilized in CRISPR/Cas-mediated cancer therapy. Cancer Gene Therapy, 2023, 30: 936-954.
Gene editing-based therapeutic strategies grant the power to override cell machinery and alter faulty genes contributing to disease development like cancer. Nowadays, the principal tool for gene editing is the clustered regularly interspaced short palindromic repeats-associated nuclease 9 (CRISPR/Cas9) system. In order to bring this gene-editing system from the bench to the bedside, a significant hurdle remains, and that is the delivery of CRISPR/Cas to various target cells in vivo and in vitro. The CRISPR-Cas system can be delivered into mammalian cells using various strategies; among all, we have reviewed recent research around two natural gene delivery systems that have been proven to be compatible with human cells. Herein, we have discussed the advantages and limitations of viral vectors, and extracellular vesicles (EVs) in delivering the CRISPR/Cas system for cancer therapy purposes.© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.
[28]
Boucher P, Cui X X, Curiel D T. Adenoviral vectors for in vivo delivery of CRISPR-Cas gene editors. Journal of Controlled Release, 2020, 327: 788-800.
Harnessing the bacterial clustered regularly interspaced short palindromic repeats (CRISPR) system for genome editing in eukaryotes has revolutionized basic biomedical research and translational sciences. The ability to create targeted alterations of the genome through this easy to design system has presented unprecedented opportunities to treat inherited disorders and other diseases such as cancer through gene therapy. A major hurdle is the lack of an efficient and safe in vivo delivery system, limiting most of the current gene therapy efforts to ex vivo editing of extracted cells. Here we discuss the unique features of adenoviral vectors that enable tissue specific and efficient delivery of the CRISPR-Cas machinery for in vivo genome editing.Copyright © 2020 Elsevier B.V. All rights reserved.
[29]
Choi J G, Dang Y, Abraham S, et al. Lentivirus pre-packed with Cas 9 protein for safer gene editing. Gene Therapy, 2016, 23(7): 627-633.
The CRISPR/Cas9 system provides an easy way to edit specific site/s in the genome and thus offers tremendous opportunity for human gene therapy for a wide range of diseases. However, one major concern is off-target effects, particularly with long-term expression of Cas9 nuclease when traditional expression methods such as via plasmid/viral vectors are used. To overcome this limitation, we pre-packaged Cas9 protein (Cas9P LV) in lentiviral particles for transient exposure and showed its effectiveness for gene disruption in cells, including primary T cells expressing specific single guide RNAs (sgRNAs). We then constructed an 'all in one virus' to express sgRNAs in association with pre-packaged Cas9 protein (sgRNA/Cas9P LV). We successfully edited CCR5 in TZM-bl cells by this approach. Using an sgRNA-targeting HIV long terminal repeat, we also were able to disrupt HIV provirus in the J-LAT model of viral latency. Moreover, we also found that pre-packaging Cas9 protein in LV particle reduced off-target editing of chromosome 4:-29134166 locus by CCR5 sgRNA, compared with continued expression from the vector. These results show that sgRNA/Cas9P LV can be used as a safer approach for human gene therapy applications.
[30]
李景虹. 多重靶向纳米递送系统为CRISPR/Cas9体内编辑提供输运工具. 科学通报, 2019, 64(25): 2561-2563.
Li J H. Multi-targeting nano-delivery system provides vehicle for in vivo CRISPR/Cas9 gene editing. Chinese Science Bulletin, 2019, 64(25): 2561-2563.
[31]
Lee K, Conboy M, Park H M, et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nature Biomedical Engineering, 2017, 1: 889-901.
[32]
Glass Z, Li Y M, Xu Q B. Nanoparticles for CRISPR-Cas9 delivery. Nature Biomedical Engineering, 2017, 1: 854-855.
The DNA mutation that causes Duchenne muscular dystrophy in mice can be corrected, with minimal off-target effects, by gold nanoparticles carrying the CRISPR components.
[33]
Liu J J, Li G M, Guo H H, et al. Dual-responsive core-shell tecto dendrimers enable efficient gene editing of cancer cells to boost immune checkpoint blockade therapy. ACS Applied Materials & Interfaces, 2023, 15(10): 12809-12821.
[34]
Li L, Hu S, Chen X Y. Non-viral delivery systems for CRISPR/Cas9-based genome editing: challenges and opportunities. Biomaterials, 2018, 171: 207-218.
In recent years, CRISPR (clustered regularly interspaced short palindromic repeat)/Cas (CRISPR-associated) genome editing systems have become one of the most robust platforms in basic biomedical research and therapeutic applications. To date, efficient in vivo delivery of the CRISPR/Cas9 system to the targeted cells remains a challenge. Although viral vectors have been widely used in the delivery of the CRISPR/Cas9 system in vitro and in vivo, their fundamental shortcomings, such as the risk of carcinogenesis, limited insertion size, immune responses and difficulty in large-scale production, severely limit their further applications. Alternative non-viral delivery systems for CRISPR/Cas9 are urgently needed. With the rapid development of non-viral vectors, lipid- or polymer-based nanocarriers have shown great potential for CRISPR/Cas9 delivery. In this review, we analyze the pros and cons of delivering CRISPR/Cas9 systems in the form of plasmid, mRNA, or protein and then discuss the limitations and challenges of CRISPR/Cas9-based genome editing. Furthermore, current non-viral vectors that have been applied for CRISPR/Cas9 delivery in vitro and in vivo are outlined in details. Finally, critical obstacles for non-viral delivery of CRISPR/Cas9 system are highlighted and promising strategies to overcome these barriers are proposed.Published by Elsevier Ltd.
[35]
Yi K, Kong H M, Zheng C X, et al. A LIGHTFUL nanomedicine overcomes EGFR-mediated drug resistance for enhanced tyrosine-kinase-inhibitor-based hepatocellular carcinoma therapy. Biomaterials, 2023, 302: 122349.
[36]
吕慧中, 赵晨辰, 朱链, 等. 外泌体靶向递药在肿瘤治疗中的进展. 中国生物工程杂志, 2021, 41(5): 79-86.
Lv H Z, Zhao C C, Zhu L, et al. Progress of using exosome for drug targeted delivery in tumor therapy. China Biotechnology, 2021, 41(5): 79-86.
[37]
Kim S M, Yang Y, Oh S J, et al. Cancer-derived exosomes as a delivery platform of CRISPR/Cas9 confer cancer cell tropism-dependent targeting. Journal of Controlled Release, 2017, 266: 8-16.
An intracellular delivery system for CRISPR/Cas9 is crucial for its application as a therapeutic genome editing technology in a broad range of diseases. Current vehicles carrying CRISPR/Cas9 limit in vivo delivery because of low tolerance and immunogenicity; thus, the in vivo delivery of genome editing remains challenging. Here, we report that cancer-derived exosomes function as natural carriers that can efficiently deliver CRISPR/Cas9 plasmids to cancer. Compared to epithelial cell-derived exosomes, cancer-derived exosomes provide potential vehicles for effective in vivo delivery via selective accumulation in ovarian cancer tumors of SKOV3 xenograft mice, most likely because of their cell tropism. CRISPR/Cas9-loaded exosomes can suppress expression of poly (ADP-ribose) polymerase-1 (PARP-1), resulting in the induction of apoptosis in ovarian cancer. Furthermore, the inhibition of PARP-1 by CRISPR/Cas9-mediated genome editing enhances the chemosensitivity to cisplatin, showing synergistic cytotoxicity. Based on these results, tumor-derived exosomes may be very promising for cancer therapeutics in the future.Copyright © 2017 Elsevier B.V. All rights reserved.
[38]
Wang L L, Wu X, Ruan Y, et al. Exosome-transmitted hsa_circ_0012634 suppresses pancreatic ductal adenocarcinoma progression through regulating miR-147b/HIPK2 axis. Cancer Biology & Therapy, 2023, 24(1): 2218514.
[39]
Wang Q Y, Yu J J, Kadungure T, et al. ARMMs as a versatile platform for intracellular delivery of macromolecules. Nature Communications, 2018, 9: 960.
Majority of disease-modifying therapeutic targets are restricted to the intracellular space and are therefore not druggable using existing biologic modalities. The ability to efficiently deliver macromolecules inside target cells or tissues would greatly expand the current landscape of therapeutic targets for future generations of biologic drugs, but remains challenging. Here we report the use of extracellular vesicles, known as arrestin domain containing protein 1 [ARRDC1]-mediated microvesicles (ARMMs), for packaging and intracellular delivery of a myriad of macromolecules, including the tumor suppressor p53 protein, RNAs, and the genome-editing CRISPR-Cas9/guide RNA complex. We demonstrate selective recruitment of these macromolecules into ARMMs. When delivered intracellularly via ARMMs, these macromolecules are biologically active in recipient cells. P53 delivered via ARMMs induces DNA damage-dependent apoptosis in multiple tissues in mice. Together, our results provide proof-of-principle demonstration that ARMMs represent a highly versatile platform for packaging and intracellular delivery of therapeutic macromolecules.
[40]
Chiang-Ni C, Chiang C Y, Chen Y W, et al. RopB-regulated SpeB cysteine protease degrades extracellular vesicles-associated streptolysin O and bacterial proteins from group A Streptococcus. Virulence, 2023, 14(1): 2249784.
[41]
Gao R R, Luo Q, Li Y, et al. Biosynthetic nanobubble-mediated CRISPR/Cas 9 gene editing of Cdh2 inhibits breast cancer metastasis. Pharmaceutics, 2022, 14(7): 1382.
[42]
Liu Y, Smid E J, Abee T, et al. Delivery of genome editing tools by bacterial extracellular vesicles. Microbial Biotechnology, 2019, 12(1): 71-73.
Here we propose to use bacterial cells as factories that generate EVs harboring both the RNP complex (or any other CRISPR-mediated tool) and a specific ligand molecule. The ligand would allow specific binding of EVs to cells with the matching receptors, adding a level of specificity to the delivery, hence stimulating precise genome editing.© 2018 The Authors. Microbial Biotechnology published by John Wiley & Sons Ltd and Society for Applied Microbiology.
[43]
Majeau N, Fortin-Archambault A, Gérard C, et al. Serum extracellular vesicles for delivery of CRISPR-CAS 9 ribonucleoproteins to modify the dystrophin gene. Molecular Therapy, 2022, 30(7): 2429-2442.
[44]
Singh A D, Patnam S, Manocha A, et al. Polyethylene glycol-based isolation of urinary extracellular vesicles, an easily adoptable protocol. MethodsX, 2023, 11: 102310.
[45]
Wan T, Zhong J F, Pan Q, et al. Exosome-mediated delivery of Cas 9 ribonucleoprotein complexes for tissue-specific gene therapy of liver diseases. Science Advances, 2022, 8(37): eabp9435.
[46]
Lakshmi S, Hughes T A, Priya S. Exosomes and exosomal RNAs in breast cancer: a status update. European Journal of Cancer, 2021, 144: 252-268.
Improved treatment of breast cancer, the world's second most common cancer, requires identification of new sensitive prognostic and diagnostic biomarkers. Exosomes are lipid-bilayer extracellular vesicles of size 30-150 nm, released by all cell types, including breast cancer cells. Cellular communication is the primary function attributed to them. This review discusses the potential utility of exosomes and exosomal RNAs (microRNAs [miRNAs]/long non-coding RNAs [LncRNAs]) in breast cancer biology and treatment. The existing literature shows that exosomes play a significant role in breast tumorigenesis and progression through transfer miRNAs and LncRNAs. These miRNAs and LncRNAs function by post-transcriptionally regulating their target mRNAs, eventually leading to modulation of expression/repression. Over the past two decades, numerous publications point towards diagnostic and therapeutic applications of exosomal miRNAs/LncRNAs. Until now, we do not have clinically approved exosome-based therapeutics. Therefore, it is high time that clinicians and cancer researchers utilise exosome's benefits through randomised clinical trials for better management of breast cancer.Copyright © 2020 Elsevier Ltd. All rights reserved.
[47]
Sun Y, Xiao W J, Yu Y, et al. Colorectal cancer-derived extracellular vesicles containing HSP 70 enhance macrophage phagocytosis by up-regulating MARCO expression. Experimental Cell Research, 2023, 426(2): 113565.
[48]
Tominaga N, Kosaka N, Ono M, et al. Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nature Communications, 2015, 6: 6716.
Tominaga, Naoomi; Kosaka, Nobuyoshi; Ono, Makiko; Katsuda, Takeshi; Yoshioka, Yusuke; Ochiya, Takahiro Natl Canc Ctr, Res Inst, Div Mol & Cellular Med, Chuo Ku, Tokyo 1040045, Japan. Tominaga, Naoomi; Nakagama, Hitoshi Univ Tokyo, Grad Sch Med, Bunkyo Ku, Tokyo 1130033, Japan. Tominaga, Naoomi Japan Soc Promot Sci, Chiyoda Ku, Tokyo 1020083, Japan. Kosaka, Nobuyoshi Univ Oxford, Dept Zool, Oxford OX1 3PS, England. Kosaka, Nobuyoshi Japan Soc Promot Sci, Res Abroad, Chiyoda Ku, Tokyo 1020083, Japan. Tamura, Kenji Natl Canc Ctr, Res Inst, Div Breast & Med Oncol, Chuo Ku, Tokyo 1040045, Japan. Lotvall, Jan Univ Gothenburg, Sahlgrenska Acad, Dept Internal Med, SE-40530 Gothenburg, Sweden. Lotvall, Jan Univ Gothenburg, Sahlgrenska Acad, Dept Resp Med & Allergol, SE-40530 Gothenburg, Sweden. Nakagama, Hitoshi Natl Canc Ctr, Res Inst, Div Canc Dev Syst, Chuo Ku, Tokyo 1040045, Japan.
[49]
Sirkisoon S R, Wong G L, Aguayo N R, et al. Breast cancer extracellular vesicles-derived miR-1290 activates astrocytes in the brain metastatic microenvironment via the FOXA2→CNTF axis to promote progression of brain metastases. Cancer Letters, 2022, 540: 215726.
[50]
He C, Jaffar Ali D, Qi Y H, et al. Engineered extracellular vesicles mediated CRISPR-induced deficiency of IQGAP1/FOXM1 reverses sorafenib resistance in HCC by suppressing cancer stem cells. Journal of Nanobiotechnology, 2023, 21(1): 154.
Sorafenib resistance poses therapeutic challenges in HCC treatment, in which cancer stem cells (CSCs) plays a crucial role. CRISPR/Cas9 can be utilized as a potential technique to overcome the drug resistance. However, a safe, efficient and target specific delivery of this platform remains challenging. Extracellular vesicles (EVs), the active components of cell to cell communication, hold promising benefits as delivery platform.Herein we report the normal epithelial cell -derived EVs engineered with HN3(HLC9-EVs) show competing tumor targeting ability. Anchoring HN3 to the membrane of the EVs through LAMP2, drastically increased the specific homing of HLC9-EVs to GPC3Huh-7 cancer cells rather than co-cultured GPC3LO2 cells. Combination therapy of HCC with sorafenib and HLC9-EVs containing sgIF to silence IQGAP1 (protein responsible for reactivation of Akt/PI3K signaling in sorafenib resistance) and FOXM1 (self-renewal transcription factor in CSCs attributed to sorafenib resistance), exhibited effective synergistic anti-cancer effect both in vitro and in vivo. Our results also showed that disruption of IQGAP1/FOXM1 resulted in the reduction of CD133 population that contribute to the stemness of liver cancer cells.By reversing sorafenib resistance using combination therapeutic approach with engineered EVs encapsulated CRISPR/Cas9 and sorafenib, our study foreshadows a path for a better, accurate, reliable and successful anti-cancer therapy in the future.© 2023. The Author(s).
[51]
Zhu X L, Gao M Y, Yang Y F, et al. The CRISPR/Cas9 system delivered by extracellular vesicles. Pharmaceutics, 2023, 15(3): 984.
[52]
Hirschenberger M, Stadler N, Fellermann M, et al. CRISPA: a non-viral, transient Cas 9 delivery system based on reengineered anthrax toxin. Frontiers in Pharmacology, 2021, 12: 770283.
[53]
Zhuang J L, Tan J Z, Wu C L, et al. Extracellular vesicles engineered with valency-controlled DNA nanostructures deliver CRISPR/Cas 9 system for gene therapy. Nucleic Acids Research, 2020, 48(16): 8870-8882.
[54]
Xu Q, Zhang Z, Zhao L, et al. Tropism-facilitated delivery of CRISPR/Cas9 system with chimeric antigen receptor-extracellular vesicles against B-cell malignancies. Journal of Controlled Release, 2020, 326: 455-467.
The CRISPR/Cas9 system is an efficient genome-editing system that has been successfully applied in the field of gene therapy. However, clinical applications of the CRISPR/Cas9 system are limited by the delivery method and safety concerns. Extracellular Vesicles (EVs) can be released from almost every type of cell, and they act as shuttles to convey molecules between cells. Here, we used EVs derived from epithelial cells as a biosafety delivery platform for the CRISPR/Cas9 system and modified the EVs with a chimeric-antigen receptor (CAR) to give them selective tropism to tumors. Compared to normal EVs, CAR-EVs accumulated in cancer tumors rapidly and released the CRISPR/Cas9 system targeting the MYC oncogene efficiently, both in vitro and in vivo. Taken together, the combination of EV and CAR was confirmed to be a novel strategy facilitating the use of natural gene therapy platforms in cancer treatment in this proof-of-concept research.Copyright © 2020 Elsevier B.V. All rights reserved.
[55]
Ye Y Y, Shi Q, Yang T, et al. In vivo visualized tracking of tumor-derived extracellular vesicles using CRISPR-Cas 9 system. Technology in Cancer Research & Treatment, 2022, 21: 15330338221085370.
[56]
Gandham S, Su X Y, Wood J, et al. Technologies and standardization in research on extracellular vesicles. Trends in Biotechnology, 2020, 38(10): 1066-1098.
Extracellular vesicles (EVs) are phospholipid bilayer membrane-enclosed structures containing RNAs, proteins, lipids, metabolites, and other molecules, secreted by various cells into physiological fluids. EV-mediated transfer of biomolecules is a critical component of a variety of physiological and pathological processes. Potential applications of EVs in novel diagnostic and therapeutic strategies have brought increasing attention. However, EV research remains highly challenging due to the inherently complex biogenesis of EVs and their vast heterogeneity in size, composition, and origin. There is a need for the establishment of standardized methods that address EV heterogeneity and sources of pre-analytical and analytical variability in EV studies. Here, we review technologies developed for EV isolation and characterization and discuss paths toward standardization in EV research.Copyright © 2020 Elsevier Ltd. All rights reserved.
[57]
Chen J C, Li P L, Zhang T Y, et al. Review on strategies and technologies for exosome isolation and purification. Frontiers in Bioengineering and Biotechnology, 2021, 9: 811971.
[58]
Zhang Y, Bi J Y, Huang J Y, et al. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. International Journal of Nanomedicine, 2020, 15: 6917-6934.
Exosomes are nano-sized small extracellular vesicles secreted by cells, carrying nucleic acids, proteins, lipids and other bioactive substances to play a role in the body's physiological and pathological processes. Compared to synthetic carriers such as liposomes and nanoparticles, the endogeneity and heterogeneity of exosomes give them extensive and unique advantages in the field of disease diagnosis and treatment. However, the storage stability, low yield, low purity, and weak targeting of exosomes limit its clinical application. For this reason, further exploration is needed to optimize the above problems and facilitate future functional studies of exosomes. In this paper, the origin, classification, preparation and characterization, storage stability and applications of exosome delivery system are summarized and discussed by searching a large number of literatures.© 2020 Zhang et al.
[59]
Le M C N, Fan Z H. Exosome isolation using nanostructures and microfluidic devices. Biomedical Materials, 2021, 16(2): 022005.
[60]
Lamparski H G, Metha-Damani A, Yao J Y, et al. Production and characterization of clinical grade exosomes derived from dendritic cells. Journal of Immunological Methods, 2002, 270(2): 211-226.
We describe methods for the production, purification, and characterization of clinical grade (cGMP) exosomes derived from antigen presenting cells (APCs). Exosomes have been shown to have immunotherapeutic properties through their presentation of biologically relevant antigens [Nat. Med. 4 (1998) 594] and are being developed as an alternative to cellular therapies. Exosomes are 50-90-nm-diameter vesicles secreted from multivesicular bodies (MVBs) found in a variety of both hematopoietic and tumor cells. These particles contain antigen presenting molecules (MHC class I, MHC class II, and CD1), tetraspan molecules (CD9, CD63, CD81), adhesion molecules (CD11b and CD54), and costimulatory molecules (CD86); hence, providing them the necessary machinery required for generating a potent immune response [J. Biol. Chem. 273 (1998) 20121; J. Cell. Sci. 113 (2000) 3365; J. Immunol. Methods 247 (2001) 163; J. Immunol. 166 (2001) 7309]. Exosomes from monocyte-derived dendritic cells (MDDCs) were rapidly purified (e.g. 4-6 h of a 2-3 l culture) based on their unique size and density. Ultrafiltration of the clarified supernatant through a 500-kDa membrane and ultracentrifugation into a 30% sucrose/deuterium oxide (D2O) (98%) cushion (density 1.210 g/cm3) reduced the volume and protein concentration approximately 200- and 1000-fold, respectively. The percentage recovery of exosomes ranged from 40% to 50% based on the exosome MHC class II concentration of the starting clarified supernatant. This methodology was extended to a miniscale process with comparable results. Conversely, the classical differential centrifugation technique is a more lengthy and variable process resulting in exosomes being contaminated with media proteins and containing only 5-25% of the starting exosome MHC class II concentration; hence, making it difficult for their use in clinical development. Lastly, we developed the following quality control assays to standardize the exosome vaccine: quantity (concentration of MHC class II) and protein characterization (FACS). The combination of a rapid and reproducible purification method and quality control assays for exosomes has allowed for its evaluation as a cancer vaccine in clinical trials [Proc. Am. Soc. Oncol. 21 (2002) 11a].
[61]
Zhu X H, Badawi M, Pomeroy S, et al. Comprehensive toxicity and immunogenicity studies reveal minimal effects in mice following sustained dosing of extracellular vesicles derived from HEK293T cells. Journal of Extracellular Vesicles, 2017, 6(1): 1324730.
[62]
Duan T Q, Gao Z L, Luo A X, et al. Adipose mesenchymal stem cell-derived extracellular vesicles reduce glutamate-induced excitotoxicity in the retina. Neural Regeneration Research, 2023, 18(10): 2315-2320.
[63]
Luo H Y, Birjandi A A, Ren F L, et al. Advances in oral mesenchymal stem cell-derived extracellular vesicles in health and disease. Genes & Diseases, 2024, 11(1): 346-357.
[64]
Luo H Y, Zhou Y H, Zhang J, et al. NK cell-derived exosomes enhance the anti-tumor effects against ovarian cancer by delivering cisplatin and reactivating NK cell functions. Frontiers in Immunology, 2022, 13: 1087689.
[65]
Bernareggi D, Xie Q, Prager B C, et al. CHMP2A regulates tumor sensitivity to natural killer cell-mediated cytotoxicity. Nature Communications, 2022, 13: 1899.
Natural killer (NK) cells are known to mediate killing of various cancer types, but tumor cells can develop resistance mechanisms to escape NK cell-mediated killing. Here, we use a "two cell type" whole genome CRISPR-Cas9 screening system to discover key regulators of tumor sensitivity and resistance to NK cell-mediated cytotoxicity in human glioblastoma stem cells (GSC). We identify CHMP2A as a regulator of GSC resistance to NK cell-mediated cytotoxicity and we confirm these findings in a head and neck squamous cells carcinoma (HNSCC) model. We show that deletion of CHMP2A activates NF-κB in tumor cells to mediate increased chemokine secretion that promotes NK cell migration towards tumor cells. In the HNSCC model we demonstrate that CHMP2A mediates tumor resistance to NK cells via secretion of extracellular vesicles (EVs) that express MICA/B and TRAIL. These secreted ligands induce apoptosis of NK cells to inhibit their antitumor activity. To confirm these in vitro studies, we demonstrate that deletion of CHMP2A in CAL27 HNSCC cells leads to increased NK cell-mediated killing in a xenograft immunodeficient mouse model. These findings illustrate a mechanism of tumor immune escape through EVs secretion and identify inhibition of CHMP2A and related targets as opportunities to improve NK cell-mediated immunotherapy.© 2022. The Author(s).
[66]
Liu X, Cao Z C, Wang W Z, et al. Engineered extracellular vesicle-delivered CRISPR/Cas 9 for radiotherapy sensitization of glioblastoma. ACS Nano, 2023, 17(17): 16432-16447.
[67]
Chen R, Yuan W L, Zheng Y J, et al. Delivery of engineered extracellular vesicles with miR-29b editing system for muscle atrophy therapy. Journal of Nanobiotechnology, 2022, 20(1): 304.
Muscle atrophy is a frequently observed complication, characterized by the loss of muscle mass and strength, which diminishes the quality of life and survival. No effective therapy except exercise is currently available. In our previous study, repressing miR-29b has been shown to reduce muscle atrophy. In our current study, we have constructed artificially engineered extracellular vesicles for the delivery of CRISPR/Cas9 to target miR-29b (EVs-Cas9-29b). EVs-Cas9-29b has shown a favorable functional effect with respect to miR-29b repression in a specific and rapid manner by gene editing. In in vitro conditions, EVs-Cas9-29b could protect against muscle atrophy induced by dexamethasone (Dex), angiotensin II (AngII), and tumor necrosis factor-alpha (TNF-α). And EVs-Cas9-29b introduced in vivo preserved muscle function in the well-established immobilization and denervation-induced muscle atrophy mice model. Our work demonstrates an engineered extracellular vesicles delivery of the miR-29b editing system, which could be potentially used for muscle atrophy therapy.© 2022. The Author(s).
[68]
Lainšček D, Kadunc L, Keber M M, et al. Delivery of an artificial transcription regulator dCas9-VPR by extracellular vesicles for therapeutic gene activation. ACS Synthetic Biology, 2018, 7(12): 2715-2725.
The CRISPR/Cas system has been developed as a potent tool for genome engineering and transcription regulation. However, the efficiency of the delivery of the system into cells, particularly for therapeutic in vivo applications, remains a major bottleneck. Extracellular vesicles (EVs), released by eukaryotic cells, can mediate the transfer of various molecules, including nucleic acids and proteins. We show the packaging and delivery of the CRISPR/Cas system via EVs to the target cells, combining the advantages of both technological platforms. A genome editing with designed extracellular vesicles (GEDEX) system generated by the producer cells can transfer the designed transcriptional regulator dCas9-VPR complexed with appropriate targeting gRNAs enabling activation of gene transcription. We show functional delivery in mammalian cells as well in the animals. The therapeutic efficiency of in vivo delivery of dCas9-VPR/sgRNA GEDEX is demonstrated in a mouse model of liver damage counteracted by upregulation of the endogenous hepatocyte growth factor, demonstrating the potential for therapeutic applications.
[69]
Zeng W J, Zheng L W, Li Y K, et al. Engineered extracellular vesicles for delivering functional Cas9/gRNA to eliminate hepatitis B virus cccDNA and integration. Emerging Microbes & Infections, 2024, 13(1): 2284286.
PDF(593 KB)

Accesses

Citation

Detail

Sections
Recommended

/