Viral Vector Delivery Techniques for CRISPR-based Homology-Directed Repair Gene Therapy with a Focus on Cystic Fibrosis

Authors

  • Sidhya Peddinti Cornell University
  • Siri Peddinti
  • Dr Michael James Essential Biotechnology LLC

DOI:

https://doi.org/10.47611/jsr.v13i2.2512

Keywords:

Gene Editing, Viral Vector Delivery, CRISPR, Gene Therapy, Homology Directed Repair, Cystic Fibrosis

Abstract

ABSTRACT

CRISPR-Cas9 is a revolutionary technology used to edit or alter an organism’s DNA sequence and gene functions. CRISPR-Cas9 technology is implemented using gene therapy vectors to effectively deliver gene editing reagents while protecting the genetic material and bypassing extracellular and cellular barriers [1]. Among current gene therapy vector technologies, viral vectors have demonstrated highly specific targeted delivery and high transduction efficiency. Viral vectors are modeled after pathogenic virus systems and integrate therapeutic genes into an organism’s genome [2]. Due to their diverse types and unique advantages and disadvantages, thorough deliberation must be applied to select an optimal classification to maintain efficacy and safety of CRISPR-Cas9 gene therapy techniques. Viral vectors can be used to effectively deliver gene therapies to repair mutations employing 2 techniques: Homology-Directed Repair (HDR) and prime editing [3, 4]. Single mutations can cause certain genetic diseases, such as cystic fibrosis, which is most commonly caused by a deletion of the 508th amino acid residue (phenylalanine) of the cystic fibrosis transmembrane conductance regulator (CFTR) protein [5, 6]. HDR repairs double-stranded breaks following the gene editing process, while prime editing, a newer technology, edits and repairs DNA without creating double-stranded breaks. These techniques are both effective options for employing CRISPR-Cas9 to repair the CFTR mutation [7, 8]. This review explores the range of available viral vector technologies used for gene delivery and compares prime editing with HDR with an emphasis on the application of repairing the CFTR deletion mutation that causes cystic fibrosis.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Author Biography

Dr Michael James, Essential Biotechnology LLC

Profile Summary:

BACKGROUND

I am an experienced scientist, consultant and writer with a background of deep expertise in medical and biological science. My PhD in microbiology was obtained in the study of the oncogenic transformation of human cells by DNA tumor viruses. My professional research career began in the study of novel oncology targets identified by genome wide association and functional genetics studies. Using molecular and cellular biology approaches as well as murine modeling, I have elucidated roles and mechanisms for several proteins involved in tumor pathobiology with potential as oncology targets.

I am a consultant for physicians, patients, academic researchers and the biotech/pharma industries. My writing and editing experience includes grant writing, scientific writing for top publishers such as Wiley and 9 years as a specialty editor for scientific content, English grammar, clarity, flow, formatting, and organization for peer review. Many of these articles have been for authors with English as a second language. I also serve as a scientific peer review and talent evaluation consultant.

As Assistant Professor of Surgical Oncology at Medical College of Wisconsin I have directed a research program that investigates the role of stress signaling in chemoresistance and identification of targetable mechanisms for re-sensitization of tumors to drugs with a focus on pancreatic cancer. In the course of my research, I have published in several high-profile journals including Cancer Research PMIDs:24366883, 19622765; 19789337; International Journal of Cancer PMID: 30468251, Oncogene PMID: 19915612, Nature Precision Oncology PMID: 33654182 Nature Genetics PMID: 21614091. In the pursuit of more informative and representative disease models, I also developed and implemented primary human pancreatic ductal adenocarcinoma organoid cultures and three-dimensional models of the PDA tumor microenvironment: PMID: 29587663. As principal investigator, my work has been funded by grants written myself and submitted to American Cancer Society, National Science Foundation, and Wellcome Trust. I am also experienced in writing SBIR and STTR grants and have been a PI on an NSF SBIR grant to develop a nanoparticle platform to deliver nucleic acid therapies to the lung and to be used to treat and prevent Covid-19.

I founded Essential Biotechnology, LLC with the goal of developing and commercializing biologic inhibitors of CRR9/CLPTM1L. This venture was based on my discovery of a role for CLPTM1L in resistance of tumor cells to killing by anoikis and by chemotherapeutic stress. We have received awards and accolades from Wellcome Trust, U.K., Licensing Executives Society International, Bridge to Cure, and SE Wisconsin Clinical and Translational Science Institute. In my role at Essential Biotechnology, I systematically developed and characterized fully human antibody inhibitors, and established preclinical data packages including MOA, activity, stability, affinity, specificity, tissue cross-reactivity, toxicity, and biodistribution data for presentation to scientific colleagues, investors, and licensing partners.

AREAS OF EXPERTISE

Oncology
Tumoroids
Organoids
Tumor Microenvironment
3D TME Modeling
Lung Cancer
Pancreatic Cancer
Ovarian Cancer
Drug Development
Targeted Therapeutics
Immuno-oncology
Antibody-Drug Conjugates
Molecular Biology
Cell Biology
Drug Resistance
Genetics
GWAS
Biomarkers
Microbiology
Immunology
Virology

JOURNAL REVIEW EXPERIENCE

I have been invited and served as a reviewer for the following peer-reviewed publications:
Dates Journal Impact Factor
2019 – Present Nature Protocols 11.334
2019 – Present EBioMedicine 6.68
2019 - Present Drug Development, Design and Therapy 3.082
2019 - Present Journal of Visualized Experiments 1.108
2018 – Present Theranostics 8.712
2018 – Present npj Precision Oncology 15.0 – 17.5 (projected)
2018 - present Cancer Biology and Therapy 2.879
2017 – Present Current Bioinformatics 1.189
2016 - Present Oncotarget 5.168 (2016)
2016 - Present Scientific Reports 4.122
2015 - Present World Journal of Gastroenterology 3.411
2016 - Present Experimental Lung Research 1.878 (2017)
2015 - Present Tumor Biology 3.650 (2016)
2015 - Present BMC Cancer 3.288 (2016)
2014 - Present Journal of Cancer Research and Clinical Oncology 3.503 (2016)
2013 - Present Molecular Carcinogenesis 4.808 (2014)
2012 – Present Annals of Human Genetics 2.211 (2014)
2009 - Present Int. J. Cancer 6.198 (2012)

RESEARCH TALENT EVALUATION EXPERIENCE

2017 Interviewer, Medical College of Wisconsin Cancer Center Director Search
2014 Interviewer, Department of Surgery Faculty Recruitment
2010 Interviewer, MCW Cancer Center Faculty Recruitment
2019 Grand Rounds Committee, MCW Department of Surgery

References or Bibliography

References

Wu, X., Kriz, A. J., & Sharp, P. A. (2014). Target specificity of the CRISPR-Cas9 system. Quantitative biology (Beijing, China), 2(2), 59–70. https://doi.org/10.1007/s40484-014-0030-x

Hsu PD, Lander ES, Zhang F. Development and applications of CRISPR-Cas9 for genome engineering. Cell. 2014 Jun 5;157(6):1262-78. doi:10.1016/j.cell.2014.05.010. PubMed Central: PMC4343198.

Komor AC, Badran AH, Liu DR. CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell. 2017 Apr 20;169(3):559. doi:10.1016/j.cell.2017.04.005. PubMed: 28431253.

B Zetsche, SE Volz, F Zhang. A split-Cas9 architecture for inducible genome editing and transcription modulation Nat Biotechnol, 33 (2015), pp. 139-142

Maule, G., Arosio, D., & Cereseto, A. (2020). Gene Therapy for Cystic Fibrosis: Progress and Challenges of Genome Editing. International journal of molecular sciences, 21(11), 3903. https://doi.org/10.3390/ijms21113903

Mention K., Santos L., Harrison P.T. Gene and base editing as a therapeutic option for cystic fibrosis-learning from other diseases. Genes. 2019;10:387. doi: 10.3390/genes10050387.

Klink D., Schindelhauer D., Laner A., Tucker T., Bebok Z., Schwiebert E.M., Boyd A.C., Scholte B.J. Gene delivery systems-Gene therapy vectors for cystic fibrosis. J. Cyst. Fibros. 2004;3:203–212. doi: 10.1016/j.jcf.2004.05.042.

Bednarski C., Tomczak K., Hövel B.V., Weber W.M., Cathomen T. Targeted integration of a super-exon into the CFTR locus leads to functional correction of a cystic fibrosis cell line model. PLoS ONE. 2016;11:1–15. doi: 10.1371/journal.pone.0161072.

DB Cox, RJ Platt, F Zhang. Therapeutic genome editing: prospects and challenges

SQ Tsai, Z Zheng, NT Nguyen, M Liebers, VV Topkar, V Thapar, et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases.

Nicklin, S. A., Wu, E., Nemerow, G. R., & Baker, A. H. (2016, December 14). The influence of adenovirus fiber structure and function on vector development for gene therapy. Retrieved from https://www.sciencedirect.com/science/article/pii/S1525001605002029

Cutting G. R. (2015). Cystic fibrosis genetics: from molecular understanding to clinical application. Nature reviews. Genetics, 16(1), 45–56. https://doi.org/10.1038/nrg3849

Sosnay PR, Salinas DB, White TB, Ren CL, Farrell PM, Raraigh KS, Girodon E, Castellani C. Applying Cystic Fibrosis Transmembrane Conductance Regulator Genetics and CFTR2 Data to Facilitate Diagnoses. J Pediatr. 2017 Feb;181S:S27-S32.e1. doi: 10.1016/j.jpeds.2016.09.063. PMID: 28129809.

Guggino WB, Stanton BA. New insights into cystic fibrosis: molecular switches that regulate CFTR. Nat Rev Mol Cell Biol. 2006 Jun;7(6):426-36. doi: 10.1038/nrm1949. PMID: 16723978.

Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P. D., Wu, X., Jiang, W., Marraffini, L. A., & Zhang, F. (2013, February 15). Multiplex genome engineering using CRISPR/Cas systems. Science. https://science.sciencemag.org/content/339/6121/819

Marquez Loza, L. I., Yuen, E. C., & McCray, P. B., Jr (2019). Lentiviral Vectors for the Treatment and Prevention of Cystic Fibrosis Lung Disease. Genes, 10(3), 218. https://doi.org/10.3390/genes10030218

Matsoukas I. G. (2020). Prime Editing: Genome Editing for Rare Genetic Diseases Without Double-Strand Breaks or Donor DNA. Frontiers in genetics, 11, 528. https://doi.org/10.3389/fgene.2020.00528

Geurts, M. H., de Poel, E., Pleguezuelos-Manzano, C., Oka, R., Carrillo, L., Andersson-Rolf, A., Boretto, M., Brunsveld, J. E., van Boxtel, R., Beekman, J. M., & Clevers, H. (2021). Evaluating CRISPR-based prime editing for cancer modeling and CFTR repair in organoids. Life science alliance, 4(10), e202000940. https://doi.org/10.26508/lsa.202000940

Liang, F., Han, M., Romanienko, P. J., & Jasin, M. (1998). Homology-directed repair is a major double-strand break repair pathway in mammalian cells. Proceedings of the National Academy of Sciences of the United States of America, 95(9), 5172–5177. https://doi.org/10.1073/pnas.95.9.5172

Nambiar, T.S., Billon, P., Diedenhofen, G. et al. Stimulation of CRISPR-mediated homology-directed repair by an engineered RAD18 variant. Nat Commun 10, 3395 (2019). https://doi.org/10.1038/s41467-019-11105-z

Lieber M. R. (2010). The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annual review of biochemistry, 79, 181–211. https://doi.org/10.1146/annurev.biochem.052308.093131

Seol JH, Shim EY, Lee SE. Microhomology-mediated end joining: Good, bad and ugly. Mutat Res. 2018 May;809:81-87. doi: 10.1016/j.mrfmmm.2017.07.002. Epub 2017 Jul 16. PMID: 28754468; PMCID: PMC6477918.

Davis, L., & Maizels, N. (2014, January 21). Homology-directed repair of DNA nicks via pathways distinct from canonical double-strand break repair. PNAS. https://www.pnas.org/content/pnas/111/10/E924.full.pdf

Li, G., Zhang, X., Zhong, C., Mo, J., Quan, R., Yang, J., Liu, D., Li, Z., Yang, H., & Wu, Z. (2017). Small molecules enhance CRISPR/Cas9-mediated homology-directed genome editing in primary cells. Scientific reports, 7(1), 8943. https://doi.org/10.1038/s41598-017-09306-x

Vidaud M, Fanen P, Martin J, Ghanem N, Nicolas S, Goossens M. Three point mutations in the CFTR gene in French cystic fibrosis patients: identification by denaturing gradient gel electrophoresis. Hum Genet. 1990 Sep;85(4):446-9. doi: 10.1007/BF02428305. PMID: 2210768.

Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, Sasaki N, Boymans S, Cuppen E, van der Ent CK, Nieuwenhuis EE, Beekman JM, Clevers H. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013 Dec 5;13(6):653-8. doi: 10.1016/j.stem.2013.11.002. PMID: 24315439.

Gaj, T., Staahl, B. T., Rodrigues, G. M. C., Limsirichai, P., Ekman, F. K., Doudna, J. A., & Schaffer, D. V. (2017). Targeted gene knock-in by homology-directed genome editing using Cas9 ribonucleoprotein and AAV donor delivery. Nucleic acids research, 45(11), e98. https://doi.org/10.1093/nar/gkx154

Wang, L., Li, F., Dang, L., Liang, C., Wang, C., He, B., Liu, J., Li, D., Wu, X., Xu, X., Lu, A., & Zhang, G. (2016). In Vivo Delivery Systems for Therapeutic Genome Editing. International journal of molecular sciences, 17(5), 626. https://doi.org/10.3390/ijms17050626

Chira, S., Jackson, C. S., Oprea, I., Ozturk, F., Pepper, M. S., Diaconu, I., Braicu, C., Raduly, L. Z., Calin, G. A., & Berindan-Neagoe, I. (2015). Progresses towards safe and efficient gene therapy vectors. Oncotarget, 6(31), 30675–30703. https://doi.org/10.18632/oncotarget.5169

Li, L., Hu, S., & Chen, X. (2018). Non-viral delivery systems for CRISPR/Cas9-based genome editing: Challenges and opportunities. Biomaterials, 171, 207–218. https://doi.org/10.1016/j.biomaterials.2018.04.031

Wold, W. S., & Toth, K. (2013). Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Current gene therapy, 13(6), 421–433. https://doi.org/10.2174/1566523213666131125095046.

Atasheva, S., & Shayakhmetov, D. M. (2022). Cytokine Responses to Adenovirus and Adenovirus Vectors. Viruses, 14(5), 888. https://doi.org/10.3390/v14050888

Sack, B. K., & Herzog, R. W. (2009). Evading the immune response upon in vivo gene therapy with viral vectors. Current opinion in molecular therapeutics, 11(5), 493–503.

Kurian, K. M., Watson, C. J., & Wyllie, A. H. (2000). Retroviral vectors. Molecular pathology : MP, 53(4), 173–176. https://doi.org/10.1136/mp.53.4.173

Coffin JM, Hughes SH, Varmus HE, editors. Retroviruses. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press; 1997. Principles of Retroviral Vector Design. Available from: https://www.ncbi.nlm.nih.gov/books/NBK19423/

Lee, C. S., Bishop, E. S., Zhang, R., Yu, X., Farina, E. M., Yan, S., Zhao, C., Zheng, Z., Shu, Y., Wu, X., Lei, J., Li, Y., Zhang, W., Yang, C., Wu, K., Wu, Y., Ho, S., Athiviraham, A., Lee, M. J., Wolf, J. M., … He, T. C. (2017). Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes & diseases, 4(2), 43–63. https://doi.org/10.1016/j.gendis.2017.04.001

Daya, S., & Berns, K. I. (2008). Gene therapy using adeno-associated virus vectors. Clinical microbiology reviews, 21(4), 583–593. https://doi.org/10.1128/CMR.00008-08

Deyle, D. R., & Russell, D. W. (2009). Adeno-associated virus vector integration. Current opinion in molecular therapeutics, 11(4), 442–447.

Siemens DR, Crist S, Austin JC, Tartaglia J, Ratliff T. Comparison of viral vectors: gene transfer efficiency and tissue specificity in a bladder cancer model. J Urol. 2003 Sep;170(3):979-84. doi: 10.1097/01.ju.0000070925.10039.23. PMID: 12913754.fƒ

Ranzani, M., Annunziato, S., Calabria, A., Brasca, S., Benedicenti, F., Gallina, P., Naldini, L., & Montini, E. (2014). Lentiviral vector-based insertional mutagenesis identifies genes involved in the resistance to targeted anticancer therapies. Molecular therapy : the journal of the American Society of Gene Therapy, 22(12), 2056–2068. https://doi.org/10.1038/mt.2014.174

Ogden PJ, Kelsic ED, Sinai S, Church GM. Comprehensive AAV capsid fitness landscape reveals a viral gene and enables machine-guided design. Science. 2019 Nov 29;366(6469):1139-1143. doi: 10.1126/science.aaw2900. PMID: 31780559; PMCID: PMC7197022.

Gutierrez-Guerrero, A., Abrey Recalde, M. J., Mangeot, P. E., Costa, C., Bernadin, O., Périan, S., Fusil, F., Froment, G., Martinez-Turtos, A., Krug, A., Martin, F., Benabdellah, K., Ricci, E. P., Giovannozzi, S., Gijsbers, R., Ayuso, E., Cosset, F. L., & Verhoeyen, E. (2021). Baboon Envelope Pseudotyped "Nanoblades" Carrying Cas9/gRNA Complexes Allow Efficient Genome Editing in Human T, B, and CD34+ Cells and Knock-in of AAV6-Encoded Donor DNA in CD34+ Cells. Frontiers in genome editing, 3, 604371. https://doi.org/10.3389/fgeed.2021.604371

Mangeot, P. E., Risson, V., Fusil, F., Marnef, A., Laurent, E., Blin, J., Mournetas, V., Massouridès, E., Sohier, T. J. M., Corbin, A., Aubé, F., Teixeira, M., Pinset, C., Schaeffer, L., Legube, G., Cosset, F. L., Verhoeyen, E., Ohlmann, T., & Ricci, E. P. (2019). Genome editing in primary cells and in vivo using viral-derived Nanoblades loaded with Cas9-sgRNA ribonucleoproteins. Nature communications, 10(1), 45. https://doi.org/10.1038/s41467-018-07845-z

Tiroille, V., Krug, A., Bokobza, E., Kahi, M., Bulcaen, M., Ensinck, M. M., Geurts, M. H., Hendriks, D., Vermeulen, F., Larbret, F., Gutierrez-Guerrero, A., Chen, Y., Van Zundert, I., Rocha, S., Rios, A. C., Medaer, L., Gijsbers, R., Mangeot, P. E., Clevers, H., Carlon, M. S., … Verhoeyen, E. (2023). Nanoblades allow high-level genome editing in murine and human organoids. Molecular therapy. Nucleic acids, 33, 57–74. https://doi.org/10.1016/j.omtn.2023.06.004

Anson D. S. (2004). The use of retroviral vectors for gene therapy-what are the risks? review of retroviral pathogenesis and its relevance to retroviral vector-mediated gene delivery. Genetic vaccines and therapy, 2(1), 9. https://doi.org/10.1186/1479-0556-2-9

Yi, Y., Noh, M. J., & Lee, K. H. (2011). Current advances in retroviral gene therapy. Current gene therapy, 11(3), 218–228. https://doi.org/10.2174/156652311795684740

Milone, M.C., O’Doherty, U. Clinical use of lentiviral vectors. Leukemia 32, 1529–1541 (2018). https://doi.org/10.1038/s41375-018-0106-0

Escors D, Breckpot K. Lentiviral vectors in gene therapy: their current status and future potential. Arch Immunol Ther Exp (Warsz). 2010;58:107–19.

RL Frock, J Hu, RM Meyers, YJ Ho, E Kii, FW Alt. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases.

P Mali, J Aach, PB Stranges, KM Esvelt, M Moosburner, S Kosuri, et al. CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol, 31 (2013), pp. 833-838

High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol, 31 (2013), pp. 822-826

Donoho, G., Jasin, M. and Berg, P. (1998). Analysis of gene targeting and intrachromosomal homologous recombination stimulated by genomic double-strand breaks in mouse embryonic stem cells. Mol. Cell. Biol. 18, 4070-4078. doi:10.1128/MCB.18.7.4070

Medina-Kauwe L. K. (2013). Development of adenovirus capsid proteins for targeted therapeutic delivery. Therapeutic delivery, 4(2), 267–277. https://doi.org/10.4155/tde.12.155.

Wang, D., Tai, P.W.L. & Gao, G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov 18, 358–378 (2019). https://doi.org/10.1038/s41573-019-0012-9.

Ramamoorth, M., & Narvekar, A. (2015). Non viral vectors in gene therapy- an overview. Journal of clinical and diagnostic research : JCDR, 9(1), GE01–GE6. https://doi.org/10.7860/JCDR/2015/10443.5394.

Hardee, C. L., Arévalo-Soliz, L. M., Hornstein, B. D., & Zechiedrich, L. (2017). Advances in Non-Viral DNA Vectors for Gene Therapy. Genes, 8(2), 65. https://doi.org/10.3390/genes8020065.

University of the Basque Country. (2017, February 14). Lipid nanoparticles for gene therapy. ScienceDaily. Retrieved October 12, 2020 from www.sciencedaily.com/releases/2017/02/170214095618.htm

Riley, M. K., & Vermerris, W. (2017). Recent Advances in Nanomaterials for Gene Delivery-A Review. Nanomaterials (Basel, Switzerland), 7(5), 94. https://doi.org/10.3390/nano7050094.

University of the Basque Country. (2017, February 14). Lipid nanoparticles for gene therapy. ScienceDaily. Retrieved October 12, 2020 from www.sciencedaily.com/releases/2017/02/170214095618.htm.

Ana del Pozo-Rodríguez, María Ángeles Solinís, Alicia Rodríguez-Gascón. Applicat-ions of lipid nanoparticles in gene therapy. European Journal of Pharmaceutics and Biopharmaceutics, 2016; 109: 184 DOI: 10.1016/j.ejpb.2016.10.016

Marangi, M., & Pistritto, G. (2018). Innovative Therapeutic Strategies for Cystic Fibrosis: Moving Forward to CRISPR Technique. Frontiers in pharmacology, 9, 396. https://doi.org/10.3389/fphar.2018.00396

Sander J.D., Joung J.K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 2014;32:347–355. doi: 10.1038/nbt.2842.

Nishiyama J., Mikuni T., Yasuda R. Virus-Mediated Genome Editing via Homology-Directed Repair in Mitotic and Postmitotic Cells in the Mammalian Brain. Neuron. 2017;96:755–768.e5. doi: 10.1016/j.neuron.2017.10.004.

Anzalone, A.V., Randolph, P.B., Davis, J.R. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019). https://doi.org/10.1038/s41586-019-1711-4.

Milone, M. C., & O'Doherty, U. (2018). Clinical use of lentiviral vectors. Leukemia, 32(7), 1529–1541.https://doi.org/10.1038/s41375-018-0106-0

Matsoukas I. G. (2020). Prime Editing: Genome Editing for Rare Genetic Diseases Without Double-Strand Breaks or Donor DNA. Frontiers in genetics, 11, 528. https://doi.org/10.3389/fgene.2020.00528

Wold, W. S., & Toth, K. (2013). Adenovirus vectors for gene therapy, vaccination and cancer gene therapy. Current gene therapy, 13(6), 421–433. https://doi.org/10.2174/1566523213666131125095046

Naehrig, S., Chao, C. M., & Naehrlich, L. (2017). Cystic Fibrosis. Deutsches Arzteblatt international, 114(33-34), 564–574. https://doi.org/10.3238/arztebl.2017.0564

Wang G. (2023). Genome Editing for Cystic Fibrosis. Cells, 12(12), 1555. https://doi.org/10.3390/cells12121555

Published

05-31-2024

How to Cite

Peddinti, S., Peddinti, S., & James, D. M. (2024). Viral Vector Delivery Techniques for CRISPR-based Homology-Directed Repair Gene Therapy with a Focus on Cystic Fibrosis. Journal of Student Research, 13(2). https://doi.org/10.47611/jsr.v13i2.2512

Issue

Section

Review Articles