A Narrative Review of the Research Advances in CRISPR/Cas9 for the Treatment of Solid Tumors
DOI: https://doi.org/10.62517/jmhs.202605105
Author(s)
Qinyu Liao
Affiliation(s)
Guangdong Country Garden School, Foshan, Guangdong, China
*Corresponding author
Abstract
The CRISPR-Cas9 gene editing technology offers a revolutionary method for addressing drug resistance and heterogeneity in solid tumors; however, its clinical use remains limited by inefficient in vivo delivery. This review systematically summarizes advances in this field, focusing on innovative strategies to overcome delivery bottlenecks. Firstly, the article outlines the mechanism of action of CRISPR system and development of novel tools like base editors and prime editors. Subsequently, it has a detailed comparison of the advantages, limitations, and latest applications of viral vectors, non-viral nanocarriers (including lipid nanoparticles, polymers, mesoporous silica, etc.), and physical delivery methods. Furthermore, the review integrates and analyzes the therapeutic exploration of CRISPR technology in six major types of solid tumors (non-small cell lung cancer, triple-negative breast cancer, colorectal cancer, hepatocellular carcinoma, glioblastoma, and pancreatic ductal adenocarcinoma). It covers various strategies which demonstrate preclinical tumor-suppressive effects. Finally, the review discusses the major challenges currently faced, such as delivery efficiency, off-target effects, and tumor heterogeneity. Additionally, future directions of intelligent delivery systems, high-fidelity editing tools, and personalized combination therapies are prospected.
Keywords
CRISPR/Cas9; Solid Tumors; Gene Editing; Drug Delivery Systems; Nanocarriers; Preclinical Studies; Tumor Microenvironment
References
[1] Hyuna Sung, Jacques Ferlay, Rebecca L Siegel, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021 May;71(3):209-249.
[2] Le Cong, F Ann Ran, David Cox, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013 Feb 15;339(6121):819-23.
[3] Eva Lentsch, Lifei Li, Susanne Pfeffer, et al. CRISPR/Cas9-Mediated Knock-Out of KrasG12D Mutated Pancreatic Cancer Cell Lines. Int J Mol Sci. 2019 Nov 14;20(22):5706.
[4] A-Rum Yoon, Soyeon Lee, Ju Hee Kim, et al. CRISPR-mediated ablation of TP53 and EGFR mutations enhances gefitinib sensitivity and anti-tumor efficacy in lung cancer. Mol Ther. 2024 Oct 2;32(10):3618-3628.
[5] Si-Wei Wang, Chao Gao, Yi-Min Zheng, et al. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. MOL CANCER. 2022;21(1):57.
[6] Minjiang Chen, Aiwu Mao, Min Xu, et al. CRISPR-Cas9 for cancer therapy: Opportunities and challenges. Cancer Lett.2019 Apr 10:447:48-55.
[7] Benjamin P Kleinstiver, Michelle S Prew, Shengdar Q Tsai, et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. 2015 Jul 23;523(7561):481-5.
[8] Bernd Zetsche, Jonathan S Gootenberg, Omar O Abudayyeh, et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. 2015 Oct 22;163(3):759-71.
[9] Nicole M Gaudelli, Alexis C Komor, Holly A Rees, et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature. 2017 Oct 25;551(7681):464–471.
[10] Andrew V Anzalone, Peyton B Randolph, Jessie R Davis, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019 Dec;576(7785):149-157.
[11] Taeyoung Koo, A-rum Yoon, Hee-yeon Cho, et al. Selective disruption of an oncogenic mutant allele by CRISPR/Cas9 induces efficient tumor regression. Nucleic Acids Res. July 2017:7897–7908.
[12] Qianqian Gao, Wenjie Ouyang, Bin Kang, et al. Selective targeting of the oncogenic KRAS G12S mutant allele by CRISPR/Cas9 induces efficient tumor regression. Theranostics. 2020 Apr 6;10(11):5137–5153.
[13] Jesse Boumelha, Andrea de Castro, Nourdine Bah, et al.CRISPR–Cas9 Screening Identifies KRAS-Induced COX2 as a Driver of Immunotherapy Resistance in Lung Cancer. Cancer Res (2024) 84 (14): 2231–2246.
[14] You Lu , Jianxin Xue, Tao Deng, et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat Med. 2020 May;26(5):732-740.
[15] Tong Wang, Tianyi Chen, Dazhao Li, et al. Core-shell vector-mediated co-delivery of CRISPR/Cas9 system and hydrophobic drugs against triple-negative breast cancer stem cells. J Control Release. 10 February 2025:1080-1091.
[16] Qiqi Mao, Peibin Wu, Haochen Li, et al. CRISPR/Cas9‑mediated EZH2 knockout suppresses the proliferation and migration of triple‑negative breast cancer cells. Oncology Letters. June 22, 2023.
[17] Rachel L. Mintz, Yeh-Hsing Lao, Chun-Wei Chi, et al. CRISPR/Cas9-mediated mutagenesis to validate the synergy between PARP1 inhibition and chemotherapy in BRCA1-mutated breast cancer cells. Bioeng Transl Med. January 2020.
[18] Meng Yang, Chen Zeng, Peiting Li, et al. Impact of CXCR4 and CXCR7 knockout by CRISPR/Cas9 on the function of triple-negative breast cancer cells. Onco Targets Ther. 17 May 2019: 3849-3858
[19]Haruna Takeda, Shiho Kataoka, Mizuho Nakayama, et al. CRISPR-Cas9–mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proc Natl Acad Sci USA. July 12, 2019:116 (31) 15635-15644.
[20] Wan C, Mahara S, Sun C, et al. Genome-scale CRISPR-Cas9 screen of Wnt/β-catenin signaling identifies therapeutic targets for colorectal cancer. Sci Adv. 19 May 2021.9761-9771.
[21] Tao Wan, Qi Pan, Liu C, et al. A Duplex CRISPR-Cas9 Ribonucleoprotein Nanomedicine for Colorectal Cancer Gene Therapy. Nano Lett. 2021, 21, 22, 9761–9771.
[22] Chune Yu, Dan Luo, Jing Yu, et al. Genome-wide CRISPR-cas9 knockout screening identifies GRB7 as a driver for MEK inhibitor resistance in KRAS mutant colon cancer. Oncogene. 2022: 191–203.
[23] Lai Wei, Derek Lee, Cheuk-Ting Law, et al.Genome-wide CRISPR/Cas9 library screening identified PHGDH as a critical driver for Sorafenib resistance in HCC. Nat Commun. 15 October 2019
[24] Yonggang Lu, Haoming Shen, Wenjie Huang, et al. Genome-scale CRISPR-Cas9 knockout screening in hepatocellular carcinoma with lenvatinib resistance. Cell Death Dis. 18 November 2021
[25] Feifan Yao, Suiqing Zhou, Ruizhi Zhang, et al. CRISPR/Cas9 screen reveals that targeting TRIM34 enhances ferroptosis sensitivity and augments immunotherapy efficacy in hepatocellular carcinoma. Cancer Lett. 2024 Jul 1:593:216935.
[26] Bing-Chen Zhang, Bang-Yue Luo, Jun-Jie Zou, et al. Co-delivery of sorafenib and CRISPR/Cas9 based on targeted core–shell hollow mesoporous organosilica nanoparticles for synergistic HCC therapy. ACS Appl Mater Interfaces. 2020, 12, 51, 57362–57372.
[27] Haohao Yin, Liping Sun, Yinying Pu, et al. Ultrasound-controlled CRISPR/Cas9 system augments sonodynamic therapy of hepatocellular carcinoma. ACS Cent Sci. 2021, 7, 12, 2049–2062
[28] Yan Zou, Xinhong Sun, Qingshan Yang, et al. Blood-brain barrier–penetrating single CRISPR-Cas9 nanocapsules for effective and safe glioblastoma gene therapy. Sci Adv. 20 Apr 2022.
[29] Graham MacLeod, Danielle A. Bozek, Nishani Rajakulendran, et al. Genome-wide CRISPR-Cas9 screens expose genetic vulnerabilities and mechanisms of temozolomide sensitivity in glioblastoma stem cells. Cell Rep. p971-986.e9April 16, 2019.
[30] Bryan D. Choi, Xiaoling Yu, Ana P. Castano, et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma. J Immunother Cancer. 14 November 2019.
[31] Xiao Liu, Zhengcong Cao, Weizhong Wang. Engineered extracellular vesicle-delivered CRISPR/Cas9 for radiotherapy sensitization of glioblastoma. ACS Nano. 2023, 17, 17, 16432–16447.
[32] Wang Jiang, Hao Li, Xiyu Liu, Jianping Zhang, Wuhu zhang, Tianjiao Li, Liang Liu, Xianjun Yu. Precise and efficient silencing of mutant KrasG12D by CRISPR-CasRx controls pancreatic cancer progression. Theranostics. 2020 Sep 16;10(25):11507–11519.
[33] Siyuan Zeng, Bin Lan, et al. CDK7 inhibition augments response to multidrug chemotherapy in pancreatic cancer. J Exp Clin Cancer Res. 2022 Aug 10;41:241.
[34] Andréa Witz, Julie Dardare, Aurélie Francois, et al. CRISPR/Cas9-mediated knock-in of BRCA1/2 mutations restores response to olaparib in pancreatic cancer cell lines. Sci Rep. 31 October 2023.