In Vivo Gene Therapy with CRISPR–Cas9: A Promising Therapeutic Strategy to Cure Inherited Diseases

Fei Li,Taorui Wang,Qiang Cheng
DOI: https://doi.org/10.1002/mef2.26
2022-01-01
MedComm – Future Medicine
Abstract:MedComm – Future MedicineVolume 1, Issue 2 e26 HIGHLIGHTOpen Access In vivo gene therapy with CRISPR–Cas9: A promising therapeutic strategy to cure inherited diseases Fei Li, Corresponding Author Fei Li [email protected] orcid.org/0000-0003-0985-2907 State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China Correspondence Fei Li, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China. Email: [email protected] Qiang Cheng, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Email: [email protected]Search for more papers by this authorTaorui Wang, Taorui Wang University Hospital and Faculty of Medicine, Macau University of Science and Technology, Macau, China Contribution: Writing - original draft (supporting)Search for more papers by this authorQiang Cheng, Corresponding Author Qiang Cheng [email protected] orcid.org/0000-0002-5213-5084 Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China Correspondence Fei Li, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China. Email: [email protected] Qiang Cheng, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Email: [email protected] Contribution: Writing - original draft (equal), Writing - review & editing (lead)Search for more papers by this author Fei Li, Corresponding Author Fei Li [email protected] orcid.org/0000-0003-0985-2907 State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China Correspondence Fei Li, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China. Email: [email protected] Qiang Cheng, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Email: [email protected]Search for more papers by this authorTaorui Wang, Taorui Wang University Hospital and Faculty of Medicine, Macau University of Science and Technology, Macau, China Contribution: Writing - original draft (supporting)Search for more papers by this authorQiang Cheng, Corresponding Author Qiang Cheng [email protected] orcid.org/0000-0002-5213-5084 Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China Correspondence Fei Li, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China. Email: [email protected] Qiang Cheng, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China. Email: [email protected] Contribution: Writing - original draft (equal), Writing - review & editing (lead)Search for more papers by this author First published: 08 December 2022 https://doi.org/10.1002/mef2.26AboutSectionsPDF ToolsRequest permissionExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onFacebookTwitterLinkedInRedditWechat A recent study published in the New England Journal of Medicine by Gillmore et al.1 demonstrated high efficacy and safety of NTLA-2001, a novel in vivo CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR associated protein 9)-mediated gene therapy, in the treatment of transthyretin (TTR) amyloidosis (Figure 1). Figure 1Open in figure viewerPowerPoint Lipid nanoparticle (LNP) enables hepatocyte gene editing by co-delivery of Cas9 (CRISPR associated protein 9) messenger RNA (mRNA) and single-guide RNA (sgRNA). (A) Formation of LNP–RNA via mixing RNA molecules and lipids with a microfluidic device. (B) Internalized LNP–RNA releases sgRNA and Cas9 mRNA that leads to the production of Cas9/sgRNA complex, resulting in specific gene editing in the nucleus. The images were created with BioRender.com. TTR amyloidosis is an autosomal dominant disorder characterized by the abnormal amyloid accumulation in the body tissue and organs. The results showed a mean reduction of 87% serum TTR protein in the participants receiving a single dose of 0.3 mg NTLA-2001 per kilogram, which should satisfy the clinical needs since such efficacy was comparable with Food and Drug Administration (FDA)-approved small interfering RNA (siRNA) medicine (Patisiran) targeted TTR.2 Although this study involves a very small number of patients, these data release a signal that in vivo gene editing represents a promising strategy to cure inherited diseases. CRISPR-Cas9 technology is currently the most widely used tool for gene editing. The system is mainly composed of Cas9 enzyme and guide RNA (gRNA). CRISPR are sequences derived from DNA fragments of bacteriophages that had previously infected the prokaryote. They are used to detect and destroy the DNA from similar bacteriophages in subsequent infections. The gRNA has two components, the CRISPR-derived RNA (crRNA) and the trans-activating crRNA (tracrRNA). The crRNA directs the Cas9 enzyme to the target DNA region, while tracrRNA links the crRNA to the Cas9 enzyme. Nowadays, single-guide RNA (sgRNA) is the most popular format for researchers compared to employ separated crRNA and tracrRNA. Cas9 is a dual RNA-guided DNA endonuclease enzyme from Streptococcus pyogenes utilizing CRISPR to detect foreign DNA and cleave it. After transfection of target cells with the CRISPR–Cas9 complex, sgRNA recognizes the target DNA region and guides the Cas9 enzyme there for editing. Then, the Cas9 enzyme cuts the target DNA, which would later be repaired by endogenous repair mechanisms to induce insertions and deletions (indels) of the target genome. Thus, gene knockout via a frameshift mutation is reached. CRISPR–Cas9 system is promising in the treatment of inherited disorders with ex vivo or in vivo approaches. The ex vivo approach edits the target genes outside the patient's body and transplants the edited cells back into the patient, of which a new generation of chimeric antigen receptor (CAR) T-cell therapy for cancer therapy via CRISPR-based editing was attractive. While in vivo approach edits the target genes inside the patient's body after delivering the CRISPR–Cas9 system into the target tissues or organs. In vivo gene editing has several advantages over ex vivo editing. For example, cell types like neurons may lose their functions or not survive after ex vivo gene editing, which could be avoided by in vivo approaches. Besides, in vivo approach could save time and cost, as well as edit the genes in multiple tissue types at the same time. Therefore, in vivo gene editing has great potential to treat multisystemic diseases. The main challenge of in vivo CRISPR–Cas9-mediated gene editing is their efficient and controlled delivery. Among the many types of vehicles, lipid nanoparticle (LNP) represents one of the best strategies (Figure 1). The advantages of LNP delivery systems include: (1) LNP could deliver different cargoes, such as mRNA, sgRNA, siRNA, plasmid, and protein; (2) advances in efficacy and safety that three LNP–RNA medicines have been approved by FDA2, 3; (3) less capacity limitation and allowed redosing compared with a viral vector (e.g., adeno-associated virus [AAV]); (4) facilitated optimization to reach tissue-selective targeting, may grant the LNP stronger affinity to target cells (organs) and higher efficiency of biomolecule delivery.4 The above advantages of LNP make it an ideal option for various gene therapies. In 2018, liver-targeted MC3-LNP was approved by FDA to deliver siRNA in the treatment of TTR amyloidosis, which marked the first approved siRNA drug in human history and the great potential of LNP vector for RNA-based therapy. This study by Gillmore et al. set another successful example of using LNP as a distributor of the CRISPR–Cas9 complex. AAVs were the popular carriers of the CRISPR–Cas9 system previously, but transduction of AAV is only efficient in specific organs such as the eye, brain, liver, and muscles.5 Here, a novel LNP is used to perform CRISPR–Cas9-based gene editing in the liver by co-delivery of Cas9 mRNA and sgRNA. The LNP consisted of ionizable lipid, phospholipid, PEGylated lipid, and cholesterol. Cas9 mRNA and TTR-specific sgRNA are packaged via the above LNP to produce NTLA-2001. After intravenous administration in patients, NTLA-2001 is opsonized by apolipoprotein E (ApoE) and transported through the systemic circulation directly into the liver, the main TTR production site, and then the cargoes are released into hepatocytes. After the generation of the Cas9 enzyme translated from mRNA, the Cas9/sgRNA complex is formed and enters into the nucleus to achieve indels of targeted DNA by the TTR-specific sgRNA, reducing functional TTR mRNA levels and TTR protein. As expected, serum TTR protein concentration was 52% and 87% lower in the patients receiving 0.1 and 0.3 mg NTLA-2001 per kilogram. Importantly, NTLA-2001 demonstrated well tolerability and safety in the study subjects. Only mild adverse events, including nausea, headache, decreased thyroxine, and rhinorrhea, occurred. No serious adverse events were observed. Liver function was within normal limits during the study. In summary, the study by Gillmore et al. provided clinical evidence for in vivo CRISPR–Cas9-mediated gene therapy of TTR amyloidosis. By knocking out the TTR gene, the administration of NTLA-2001 led to a significant reduction of serum TTR protein. Moreover, NTLA-2001 had high safety and only caused mild adverse events. The sample size of this study is relatively small, and the follow-up duration is limited to 28 days. Therefore, a long-term follow-up on a larger cohort of TTR amyloidosis patients is required to confirm the durability of the therapeutic effect and safety. Nonetheless, this study gives a positive ethos for gene therapy in inherited disorders via the LNP–RNA system. Looking forward to witnessing further investigation and more relative applications in the future. AUTHOR CONTRIBUTIONS Fei Li: Writing – original draft (equal); writing – review and editing. Taorui Wang: Writing – original draft (supporting). Qiang Cheng: Writing – original draft (equal); writing – review and editing (lead). All authors have read and approved the article. ACKNOWLEDGMENT We thank the support from Prof. Tuo Wei of Institute of Zoology Chinese Academy of Sciences for preparing the images for this manuscript. CONFLICT OF INTEREST The authors declare no conflict of interest. ETHICS STATEMENT Not applicable. Open Research DATA AVAILABILITY STATEMENT Not applicable. REFERENCES 1Gillmore JD, Gane E, Taubel J, et al. CRISPR–Cas9 in vivo gene editing for transthyretin amyloidosis. N Engl J Med. 2021; 385: 493- 502. 2Adams D, Gonzalez-Duarte A, O′Riordan WD, et al. Patisiran, an RNAi therapeutic, for hereditary transthyretin amyloidosis. N Engl J Med. 2018; 379(1): 11- 21. 3Schoenmaker L, Witzigmann D, Kulkarni JA, et al. mRNA–lipid nanoparticle COVID-19 vaccines: structure and stability. Int J Pharm. 2021; 601:120586. 4Cheng Q, Wei T, Farbiak L, Johnson LT, Dilliard SA, Siegwart DJ. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR–Cas gene editing. Nat Nanotechnol. 2020; 15(4): 313- 320. 5Kotterman MA, Schaffer DV. Engineering adeno-associated viruses for clinical gene therapy. Nat Rev Genet. 2014; 15(7): 445- 451. Volume1, Issue2September 2022e26 FiguresReferencesRelatedInformation
What problem does this paper attempt to address?