Abstract
Gene editing technologies help identify the genetic perturbations driving tumour initiation, growth, metastasis and resistance to therapeutics. This wealth of information highlights tumour complexity and is driving cancer research towards precision medicine approaches based on an individual’s tumour genetics. Bladder cancer is the 11th most common cancer in the UK, with high rates of relapse and low survival rates in patients with muscle-invasive bladder cancer (MIBC). MIBC is highly heterogeneous and encompasses multiple molecular subtypes, each with different responses to therapeutics. This evidence highlights the need to identify innovative therapeutic targets to address the challenges posed by this heterogeneity. CRISPR–Cas9 technologies have been used to advance our understanding of MIBC and determine novel drug targets through the identification of drug resistance mechanisms, targetable cell-cycle regulators, and novel tumour suppressor and oncogenes. However, the use of these technologies in the clinic remains a substantial challenge and will require careful consideration of dosage, safety and ethics. CRISPR–Cas9 offers considerable potential for revolutionizing bladder cancer therapies, but substantial research is required for validation before these technologies can be used in the clinical setting.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Gaj, T., Gersbach, C. A. & Barbas, C. F. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. 31, 397–405 (2013).
Xiao-Jie, L., Hui-Ying, X., Zun-Ping, K., Jin-Lian, C. & Li-Juan, J. CRISPR-Cas9: A new and promising player in gene therapy. J. Med. Genet. 52, 289–296 (2015).
Awwad, S. W., Serrano-Benitez, A., Thomas, J. C., Gupta, V. & Jackson, S. P. Revolutionizing DNA repair research and cancer therapy with CRISPR–Cas screens. Nat. Rev. Mol. Cell Biol. 24, 477–494 (2023).
Wang, M., Chen, M., Wu, X., Huang, X. & Yu, B. CRISPR applications in cancer diagnosis and treatment. Cell. Mol. Biol. Lett. 28, 73 (2023).
Das, S., Bano, S., Kapse, P. & Kundu, G. C. CRISPR based therapeutics: a new paradigm in cancer precision medicine. Molecular Cancer 21, 85 (2022).
Cancer Research UK. Bladder cancer incidence statistics. CRUK www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/bladder-cancer/incidence (2022).
Chang, S. S. et al. Diagnosis and treatment of non-muscle invasive bladder cancer: AUA/SUO Guideline. J. Urol. 196, 1021–1029 (2016).
Nadal, R. & Bellmunt, J. Management of metastatic bladder cancer. Cancer Treat. Rev. 76, 10–21 (2019).
Perera, J. & Hoskin, P. The role of radiotherapy in metastatic bladder cancer. J. Cancer Metastasis Treat. 8, 4 (2022).
Song, Y. P. et al. Long-term outcomes of radical radiation therapy with hypoxia modification with biomarker discovery for stratification: 10-year update of the BCON (Bladder Carbogen Nicotinamide) phase 3 randomized trial (ISRCTN45938399). Int. J. Radiat. Oncol. Biol. Phys. 110, 1407–1415 (2021).
Stenehjem, D. D., Tran, D., Nkrumah, M. A. & Gupta, S. PD1/PDL1 inhibitors for the treatment of advanced urothelial bladder cancer. Onco Targets Ther. 11, 5973–5989 (2018).
Lopez-Beltran, A. et al. Immune checkpoint inhibitors for the treatment of bladder cancer. Cancers 13, 131 (2021).
Massari, F. et al. Immune checkpoint inhibitors for metastatic bladder cancer. Cancer Treat. Rev. 64, 11–20 (2018).
Fong, M. H. Y., Feng, M., McConkey, D. J. & Choi, W. Update on bladder cancer molecular subtypes. Transl Androl. Urol. 9, 2881 (2020).
McConkey, D. J. & Choi, W. Molecular subtypes of bladder cancer. Curr. Oncol. Rep. 20, 1–7 (2018).
Kamoun, A. et al. A consensus molecular classification of muscle-invasive bladder cancer. Eur. Urol. 77, 420–433 (2020).
Marraffini, L. A. in Streptococcus pyogenes: Basic Biology to Clinical Manifestations (eds Ferretti, J. J., Stevens, D. L. & Fischetti V. A.) (Univ. Oklahoma Health Scinces Center, 2016).
Doudna, J. A. & Charpentier, E. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096, (2014).
Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).
Gunitseva, N., Evteeva, M., Borisova, A., Patrushev, M. & Subach, F. RNA-dependent RNA targeting by CRISPR-Cas systems: characterizations and applications. Int. J. Mol. Sci. 24, 6894 (2023).
Pickar-Oliver, A. & Gersbach, C. A. The next generation of CRISPR–Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 20, 490–507 (2019).
Hefferin, M. L. & Tomkinson, A. E. Mechanism of DNA double-strand break repair by non-homologous end joining. DNA Repair 4, 639648 (2005).
Chu, V. T. et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543–548 (2015).
Scully, R., Panday, A., Elango, R. & Willis, N. A. DNA double-strand break repair-pathway choice in somatic mammalian cells. Nat. Rev. Mol. Cell Biol. 20, 698–714 (2019).
Jensen, T. I. et al. Targeted regulation of transcription in primary cells using CRISPRa and CRISPRi. Genome Res. 31, 2120–2130 (2021).
Maeder, M. L. et al. CRISPR RNA-guided activation of endogenous human genes. Nat. Methods 10, 977–979 (2013).
Armando Casas-Mollano, J., Zinselmeier, M. H., Erickson, S. E. & Smanski, M. J. CRISPR-Cas activators for engineering gene expression in higher eukaryotes. CRISPR J. 3, 350–364 (2020).
Larson, M. H. et al. CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat. Protoc. 8, 2180–2196 (2013).
Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 422–451 (2013).
Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420–424 (2016).
Gaudelli, N. M. et al. Programmable base editing of T to G C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).
Eid, A., Alshareef, S. & Mahfouz, M. M. CRISPR base editors: Genome editing without double-stranded breaks. Biochem. J. 475, 1955–1964 (2018).
Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149–157 (2019).
Nelson, J. W. et al. Engineered pegRNAs improve prime editing efficiency. Nat. Biotechnol. 40, 402–410 (2022).
Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).
Wang, T., Wei, J. J., Sabatini, D. M. & Lander, E. S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).
Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 1–12 (2014).
Zetsche, B. et al. Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell 163, 759–771 (2015).
Zhang, C. et al. Structural Basis for the RNA-Guided Ribonuclease Activity of CRISPR-Cas13d. Cell 175, 212–223 (2018).
Gao, Y. et al. Complex transcriptional modulation with orthogonal and inducible dCas9 regulators. Nat. Methods 13, 1043–1049 (2016).
Liu, J. et al. Delivery of RIPK4 small interfering RNA for bladder cancer therapy using natural halloysite nanotubes. Sci. Adv. 5, eaaw6499 (2019).
Chen, S. H. & Zhaori, G. Potential clinical applications of siRNA technique: Benefits and limitations. Eur. J. Clin. Invest. 41, 221–232 (2011).
Isazadeh, H. et al. Advances in siRNA delivery approaches in cancer therapy: challenges and opportunities. Mol. Biol. Rep. 50, 9529–9543 (2023).
Massari, F. et al. Emerging concepts on drug resistance in bladder cancer: Implications for future strategies. Crit. Rev. Oncol. Hematol. 96, 81–90 (2015).
Galluzzi, L. et al. Molecular mechanisms of cisplatin resistance. Oncogene 31, 1869–1883 (2012).
Yu, H. M. & Wang, T. C. Mechanism of cisplatin resistance in human urothelial carcinoma cells. Food Chem. Toxicol. 50, 1226–1237 (2012).
Liu, S., Chen, X. & Lin, T. Emerging strategies for the improvement of chemotherapy in bladder cancer: Current knowledge and future perspectives. J. Adv. Res. 39, 187–202 (2022).
Kumar, G. et al. Genome-wide CRISPR screen reveals SLFN11 as a potent mediator of cisplatin sensitivity in muscle-invasive bladder cancer. Urol. Oncol. Semin. Ori. 38, 905 (2020).
Luan, J., Gao, X., Hu, F., Zhang, Y. & Gou, X. SLFN11 is a general target for enhancing the sensitivity of cancer to chemotherapy (DNA-damaging agents). J. Drug Target. 28, 33–40 (2020).
Zhang, B. et al. A wake-up call for cancer DNA damage: the role of Schlafen 11 (SLFN11) across multiple cancers. Br. J. Cancer 125, 1333–1340 (2021).
Zhang, H. et al. CircLIFR synergizes with MSH2 to attenuate chemoresistance via MutSα/ATM-p73 axis in bladder cancer. Mol. Cancer 20, 1–20 (2021).
Shi, Z.D. et al. Targeting HNRNPU to overcome cisplatin resistance in bladder cancer. Mol. Cancer 21, 37 (2022).
Lan, L. et al. Functional and physical interactions between ERCC1 and MSH2 complexes for resistance to cis-diamminedichloroplatinum(II) in mammalian cells. DNA Repair 3, 135–143 (2004).
Hoskin, P. J. et al. Carbogen and nicotinamide in the treatment of bladder cancer with radical radiotherapy. Br. J. Cancer 76, 260–263 (1997).
Hoskin, P. J., Rojas, A. M., Bentzen, S. M. & Saunders, M. I. Radiotherapy with concurrent carbogen and nicotinamide in bladder carcinoma. J. Clin. Oncol. 28, 4912–4918 (2010).
Rhea, L. P., Mendez-Marti, S., Kim, D. & Aragon-Ching, J. B. Role of immunotherapy in bladder cancer. Cancer Treat. Res. Commun. 26, 100296 (2021).
Galsky, M. D. et al. Gemcitabine and cisplatin plus nivolumab as organ-sparing treatment for muscle-invasive bladder cancer: a phase 2 trial. Nat. Med. 29, 2825–2834 (2023).
Zhang, W. et al. Disruption of CTLA-4 expression on peripheral blood CD8 + T cell enhances anti-tumor efficacy in bladder cancer. Cancer Chemother. Pharmacol. 83, 911–920 (2019).
Zhen, S. et al. Inhibition of long non-coding RNA UCA1 by CRISPR/Cas9 attenuated malignant phenotypes of bladder cancer. Oncotarget 8, 9634 (2017).
Zhen, S., Lu, J., Chen, W., Zhao, L. & Li, X. Synergistic antitumor effect on bladder cancer by rational combination of programmed cell death 1 blockade and CRISPR-Cas9-mediated long non-coding RNA urothelial carcinoma associated 1 knockout. Hum. Gene Ther. 29, 1352–1363 (2018).
Nandwani, A., Rathore, S. & Datta, M. LncRNAs in cancer: Regulatory and therapeutic implications. Cancer Lett. 501, 162–171 (2021).
He, Z. X. et al. Current development of CBP/p300 inhibitors in the last decade. Eur. J. Med. Chem. 209, 112861 (2021).
Liu, Y. et al. Synthesizing and gate genetic circuits based on CRISPR-Cas9 for identification of bladder cancer cells. Nat. Commun. 5, 5393 (2014).
Li, J. et al. A CRISPR interference of CBP and p300 selectively induced synthetic lethality in bladder cancer cells in vitro. Int. J. Biol. Sci. 15, 1276 (2019).
Zhang, M. et al. CDK inhibitors in cancer therapy, an overview of recent development. Am. J. Cancer Res. 11, 1913 (2021).
Rose, T. L. et al. Phase II trial of palbociclib in patients with metastatic urothelial cancer after failure of first-line chemotherapy. Br. J. Cancer 119, 801–807 (2018).
Tong, Z. et al. Functional genomics identifies predictive markers and clinically actionable resistance mechanisms to CDK4/6 inhibition in bladder cancer. J. Exp. Clin. Cancer Res. 38, 1–14 (2019).
He, Z., He, J. & Xie, K. KLF4 transcription factor in tumorigenesis. Cell Death Discov. 9, 118 (2023).
Xu, X. et al. CRISPR-ON-Mediated KLF4 overexpression inhibits the proliferation, migration and invasion of urothelial bladder cancer in vitro and in vivo. Oncotarget 8, 10278 (2017).
Chen, L. J. et al. The role of lysine-specific demethylase 6 A (KDM6A) in tumorigenesis and its therapeutic potentials in cancer therapy. Bioorg. Chem. 133, 106409 (2023).
Matar, M. et al. Implication of KDM6A in bladder cancer. Pharmacogenomics 24, 509–522 (2023).
Ahn, J. et al. Target sequencing and CRISPR/Cas editing reveal simultaneous loss of UTX and UTY in urothelial bladder cancer. Oncotarget 7, 63252 (2016).
Ler, L. D. et al. Loss of tumor suppressor KDM6A amplifies PRC2-regulated transcriptional repression in bladder cancer and can be targeted through inhibition of EZH2. Sci. Transl Med. 9, eaai8312 (2017).
Lang, A. et al. Contingencies of UTX/KDM6A action in urothelial carcinoma. Cancers 11, 481 (2019).
Zhuang, C. et al. Engineered CRISPR/Cas13d Sensing hTERT Selectively Inhibits the Progression of Bladder Cancer In Vitro. Front. Mol. Biosci. 8, 646412 (2021).
Hillary, V. E. & Ceasar, S. A. A review on the mechanism and applications of CRISPR/Cas9/Cas12/Cas13/Cas14 proteins utilized for genome engineering. Mol. Biotechnol. 65, 311–325 (2023).
Zhang, R., Xia, L. Q., Lu, W. W., Zhang, J. & Zhu, J. S. LncRNAs and cancer. Oncol. Lett. 12, 1233–1239 (2016).
Xie, J. et al. LncRNA SNHG3 enhances BMI1 mRNA stability by binding and regulating c-MYC: Implications for the carcinogenic role of SNHG3 in bladder cancer. Cancer Med. 12, 5718–5735 (2023).
Tortora, D. et al. A genome-wide CRISPR screen maps endogenous regulators of PPARG gene expression in bladder cancer. iScience 26, 106525 (2023).
Frangoul, H. et al. Safety and efficacy of CTX001 in patients with transfusion-dependent β-thalassemia and sickle cell disease: early results from the Climb THAL-111 and Climb SCD-121 studies of autologous CRISPR-CAS9-modified CD34+ hematopoietic stem and progenitor cells. Blood 136 (Suppl. 1), 3–4 (2020).
Hu, Y. et al. CRISPR/Cas9-engineered universal CD19/CD22 dual-targeted CAR-T cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia. Clin. Cancer Res. 27, 2764–2772 (2021).
Tsimberidou, A. M. et al. T-cell receptor-based therapy: an innovative therapeutic approach for solid tumors. J. Hematol. Oncol. 14, 102 (2021).
Tan, E., Gakhar, N. & Kirtane, K. TCR gene-engineered cell therapy for solid tumors. Best Prac. Res. Clin. Haematol. 34, 101285 (2021).
He, J. et al. Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response. Cell Res. 32, 530–542 (2022).
Foy, S. P. et al. Non-viral precision T cell receptor replacement for personalized cell therapy. Nature 615, 687–696 (2023).
Wachsmann, T. L. A. et al. Comparing CAR and TCR engineered T cell performance as a function of tumor cell exposure. Oncoimmunology 11, 2033528 (2022).
Zhang, D. et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nat. Nanotechnol. 17, 777–787 (2022).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04426669 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT05795595 (2024).
Jardim, D. L., Goodman, A., de Melo Gagliato, D. & Kurzrock, R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell 39, 154–173 (2021).
Yang, Y., Xu, J., Ge, S. & Lai, L. CRISPR/Cas: advances, limitations, and applications for precision cancer research. Front. Med. 8, 649896 (2021).
Wang, S. W. et al. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol. Cancer 21, 57 (2022).
Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187–197 (2015).
Gillmore, J. D. et al. CRISPR-Cas9 in vivo gene editing for transthyretin amyloidosis. N. Engl. J. Med. 385, 493–502 (2021).
Aimo, A. et al. RNA-targeting and gene editing therapies for transthyretin amyloidosis. Nat. Rev. Cardiol. 19, 655–667 (2022).
Sýkora, P. Germline Gene Therapy in the Era of Precise Genome Editing: How Far Should We Go? in The Ethics of Reproductive Genetics Philosophy and Medicine Ch. 11 (ed. Soniewicka, M.) 157-171 (Springer, 2018).
Jiang, F. & Doudna, J. A. CRISPR-Cas9 Structures and Mechanisms. Annu. Rev. Biophys. 46, 505–529 (2017).
Brokowski, C. & Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J. Mol. Biol. 431, 88–101 (2019).
Dang, Y. et al. Optimizing sgRNA structure to improve CRISPR-Cas9 knockout efficiency. Genome Biol. 16, 1–10 (2015).
Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62–67 (2014).
Gong, S., Yu, H. H., Johnson, K. A. & Taylor, D. W. DNA unwinding is the primary determinant of CRISPR-Cas9 activity. Cell Rep. 22, 359–371 (2018).
Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).
Davis, A. J. & Chen, D. J. DNA double strand break repair via non-homologous end-joining. Transl Cancer Res. 2, 130 (2013).
Liang, F., Han, M., Romanienko, P. J. & Jasin, M. Homology-directed repair is a major double-strand break repair pathway in mammalian cells. Proc. Natl Acad. Sci. USA 95, 5172–5177 (1998).
Lino, C. A., Harper, J. C., Carney, J. P. & Timlin, J. A. Delivering CRISPR: A review of the challenges and approaches. Drug Deliv. 25, 1234–1257 (2018).
Author information
Authors and Affiliations
Contributions
D.J.S. researched data for the article. D.J.S. contributed substantially to discussion of the content. D.J.S. wrote the article. S.L., A.D.A., S.N., T.A.D.S., K.J.R., P.J.H. and A.C. reviewed and/or edited the manuscript before submission.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Urology thanks Michele Hoffmann, Yu-chen Liu, Mads Daugaard and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Smith, D.J., Lunj, S., Adamson, A.D. et al. CRISPR–Cas9 potential for identifying novel therapeutic targets in muscle-invasive bladder cancer. Nat Rev Urol 22, 55–65 (2025). https://doi.org/10.1038/s41585-024-00901-y
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41585-024-00901-y