1
|
Mishra G, Srivastava K, Rais J, Dixit M, Kumari Singh V, Chandra Mishra L. CRISPR-Cas9: A Potent Gene-editing Tool for the Treatment of Cancer. Curr Mol Med 2024; 24:191-204. [PMID: 36788695 DOI: 10.2174/1566524023666230213094308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 11/02/2022] [Accepted: 11/10/2022] [Indexed: 02/16/2023]
Abstract
The prokaryotic adaptive immune system has clustered regularly interspaced short palindromic repeat. CRISPR-associated protein (CRISPR-Cas) genome editing systems have been harnessed. A robust programmed technique for efficient and accurate genome editing and gene targeting has been developed. Engineered cell therapy, in vivo gene therapy, animal modeling, and cancer diagnosis and treatment are all possible applications of this ground-breaking approach. Multiple genetic and epigenetic changes in cancer cells induce malignant cell growth and provide chemoresistance. The capacity to repair or ablate such mutations has enormous potential in the fight against cancer. The CRISPR-Cas9 genome editing method has recently become popular in cancer treatment research due to its excellent efficiency and accuracy. The preceding study has shown therapeutic potential in expanding our anticancer treatments by using CRISPR-Cas9 to directly target cancer cell genomic DNA in cellular and animal cancer models. In addition, CRISPR-Cas9 can combat oncogenic infections and test anticancer medicines. It may design immune cells and oncolytic viruses for cancer immunotherapeutic applications. In this review, these preclinical CRISPRCas9- based cancer therapeutic techniques are summarised, along with the hurdles and advancements in converting therapeutic CRISPR-Cas9 into clinical use. It will increase their applicability in cancer research.
Collapse
Affiliation(s)
- Gauri Mishra
- Department of Zoology, Swami Shraddhanand College, University of Delhi-110036, Delhi, India
- Division Radiopharmaceuticals and Radiation Biology, Institute of Nuclear Medicine and Allied Sciences, Brig SK Mazumdar Road, Delhi-110054, India
| | - Kamakshi Srivastava
- Department of Zoology, Swami Shraddhanand College, University of Delhi-110036, Delhi, India
| | - Juhi Rais
- Department of Nuclear Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow-226014, India
| | - Manish Dixit
- Department of Nuclear Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow-226014, India
| | - Vandana Kumari Singh
- Department of Zoology, Hansraj College, University of Delhi- 110007, Dehli, India
| | | |
Collapse
|
2
|
Kumar A, Das SK, Emdad L, Fisher PB. Applications of tissue-specific and cancer-selective gene promoters for cancer diagnosis and therapy. Adv Cancer Res 2023; 160:253-315. [PMID: 37704290 DOI: 10.1016/bs.acr.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Current treatment of solid tumors with standard of care chemotherapies, radiation therapy and/or immunotherapies are often limited by severe adverse toxic effects, resulting in a narrow therapeutic index. Cancer gene therapy represents a targeted approach that in principle could significantly reduce undesirable side effects in normal tissues while significantly inhibiting tumor growth and progression. To be effective, this strategy requires a clear understanding of the molecular biology of cancer development and evolution and developing biological vectors that can serve as vehicles to target cancer cells. The advent and fine tuning of omics technologies that permit the collective and spatial recognition of genes (genomics), mRNAs (transcriptomics), proteins (proteomics), metabolites (metabolomics), epiomics (epigenomics, epitranscriptomics, and epiproteomics), and their interactomics in defined complex biological samples provide a roadmap for identifying crucial targets of relevance to the cancer paradigm. Combining these strategies with identified genetic elements that control target gene expression uncovers significant opportunities for developing guided gene-based therapeutics for cancer. The purpose of this review is to overview the current state and potential limitations in developing gene promoter-directed targeted expression of key genes and highlights their potential applications in cancer gene therapy.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
3
|
Telomerase gene therapy: a remission toward cancer. Med Oncol 2022; 39:105. [DOI: 10.1007/s12032-022-01702-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/02/2022] [Indexed: 12/21/2022]
|
4
|
Zhuang C, Zhuang C, Zhou Q, Huang X, Gui Y, Lai Y, Yang S. Engineered CRISPR/Cas13d Sensing hTERT Selectively Inhibits the Progression of Bladder Cancer In Vitro. Front Mol Biosci 2021; 8:646412. [PMID: 33816560 PMCID: PMC8017217 DOI: 10.3389/fmolb.2021.646412] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Aptazyme and CRISPR/Cas gene editing system were widely used for regulating gene expression in various diseases, including cancer. This work aimed to reconstruct CRISPR/Cas13d tool for sensing hTERT exclusively based on the new device OFF-switch hTERT aptazyme that was inserted into the 3’ UTR of the Cas13d. In bladder cancer cells, hTERT ligand bound to aptamer in OFF-switch hTERT aptazyme to inhibit the degradation of Cas13d. Results showed that engineered CRISPR/Cas13d sensing hTERT suppressed cell proliferation, migration, invasion and induced cell apoptosis in bladder cancer 5637 and T24 cells without affecting normal HFF cells. In short, we constructed engineered CRISPR/Cas13d sensing hTERT selectively inhibited the progression of bladder cancer cells significantly. It may serve as a promising specifically effective therapy for bladder cancer cells.
Collapse
Affiliation(s)
- Chengle Zhuang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Changshui Zhuang
- Department of Urology, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen, China
| | - Qun Zhou
- Department of Urology, the Affiliated Nanhua Hospital of University of South China, Hengyang, China
| | - Xueting Huang
- Department of Nephrorheumatology, Shenzhen Yantian District People's Hospital, Shenzhen, China
| | - Yaoting Gui
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shangqi Yang
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
5
|
Seimiya H. Crossroads of telomere biology and anticancer drug discovery. Cancer Sci 2020; 111:3089-3099. [PMID: 32579791 PMCID: PMC7469838 DOI: 10.1111/cas.14540] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The telomere is the specialized nucleoprotein complex at the end of the chromosome. Its highly conserved 5'-TTAGGG-3' repeats and shelterin protein complexes form a protective loop structure to maintain the integrity and stability of linear chromosomes. Although human somatic cells gradually shorten telomeres to undergo senescence or crisis, cancer cells activate telomerase, or the recombination-based mechanism to maintain telomeres and exhibit immortality. As the most frequent non-coding mutations in cancer, gain-of-function mutations in the promoter region of the telomerase catalytic subunit, TERT, trigger telomerase activation. Promoter methylation and copy number gain are also associated with the enhanced TERT expression. Although telomerase inhibitors were pioneered from telomere-directed therapeutics, their efficacies are limited to cancer with short telomeres and some hematological malignancies. Other therapeutic approaches include a nucleoside analog incorporated to telomeres and TERT promoter-driven oncolytic adenoviruses. Tankyrase poly(ADP-ribose) polymerase, a positive regulator of telomerase, has been rediscovered as a target for Wnt-driven cancer. Meanwhile, telomeric nucleic acids form a higher-order structure called a G-quadruplex (G4). G4s are formed genome-wide and their dynamics affect various events, including replication, transcription, and translation. G4-stabilizing compounds (G4 ligands) exert anticancer effects and are in clinical investigations. Collectively, telomere biology has provided clues for deeper understanding of cancer, which expands opportunities to discover innovative anticancer drugs.
Collapse
Affiliation(s)
- Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
6
|
Telomere Gene Therapy: Polarizing Therapeutic Goals for Treatment of Various Diseases. Cells 2019; 8:cells8050392. [PMID: 31035374 PMCID: PMC6563133 DOI: 10.3390/cells8050392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Modulation of telomerase maintenance by gene therapy must meet two polarizing requirements to achieve different therapeutic outcomes: Anti-aging/regenerative applications require upregulation, while anticancer applications necessitate suppression of various genes integral to telomere maintenance (e.g., telomerase, telomerase RNA components, and shelterin complex). Patients suffering from aging-associated illnesses often exhibit telomere attrition, which promotes chromosomal instability and cellular senescence, thus requiring the transfer of telomere maintenance-related genes to improve patient outcomes. However, reactivation and overexpression of telomerase are observed in 85% of cancer patients; this process is integral to cancer immortality. Thus, telomere-associated genes in the scope of cancer gene therapy must be inactivated or inhibited to induce anticancer effects. These contradicting requirements for achieving different therapeutic outcomes mean that any vector-mediated upregulation of telomere-associated genes must be accompanied by rigorous evaluation of potential oncogenesis. Thus, this review aims to discuss how telomere-associated genes are being targeted or utilized in various gene therapy applications and provides some insight into currently available safety hazard assessments.
Collapse
|
7
|
Phelps MP, Yang H, Patel S, Rahman MM, McFadden G, Chen E. Oncolytic Virus-Mediated RAS Targeting in Rhabdomyosarcoma. MOLECULAR THERAPY-ONCOLYTICS 2018; 11:52-61. [PMID: 30364635 PMCID: PMC6197336 DOI: 10.1016/j.omto.2018.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Abstract
Aberrant activation of the receptor tyrosine kinase-mediated RAS signaling cascade is the primary driver of embryonal rhabdomyosarcoma (ERMS), a pediatric cancer characterized by a block in myogenic differentiation. To investigate the cellular function of activated RAS signaling in regulating the growth and differentiation of ERMS cells, we genetically ablated activated RAS oncogenes with high-efficiency genome-editing technology. Knockout of NRAS in CRISPR-inducible ERMS xenograft models resulted in near-complete tumor regression through a combination of cell death and myogenic differentiation. Utilizing this strategy for therapeutic RAS targeting in ERMS, we developed a recombinant oncolytic myxoma virus (MYXV) engineered with CRISPR/Cas9 gene-editing capability. Treatment of pre-clinical human ERMS tumor xenografts with an NRAS-targeting version of this MYXV significantly reduced tumor growth and increased overall survival. Our data suggest that targeted gene-editing cancer therapies have promising translational applications, especially with improvements to gene-targeting specificity and oncolytic vector technology.
Collapse
Affiliation(s)
- Michael P Phelps
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Heechang Yang
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shivani Patel
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Masmudur M Rahman
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85281, USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ 85281, USA
| | - Eleanor Chen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Peng L, Pan P, Chen J, Yu X, Wu J, Chen Y. A tetracycline-inducible CRISPR/Cas9 system, targeting two long non-coding RNAs, suppresses the malignant behavior of bladder cancer cells. Oncol Lett 2018; 16:4309-4316. [PMID: 30214566 PMCID: PMC6126189 DOI: 10.3892/ol.2018.9157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 06/28/2018] [Indexed: 12/12/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) associated protein 9 (Cas9) technology has been applied in varied biological studies, including cancer studies. However, stable mRNA expression of Cas9 has potential risks in future gene therapy. Therefore, in the present study, a tetracycline-inducible switch was used to control the mRNA expression of Cas9. Long non-coding RNAs (lncRNAs) may be important functional regulators in tumor development, including in bladder cancer. RNA was designed to simultaneously target two lncRNAs, PVT1 and ANRIL, which are considered to be bladder cancer oncogenes. The mRNA expression of Cas9 was controlled by doxycycline. Reverse transcription-quantitative polymerase chain reaction revealed that the expression of PVT1 and ANRIL was significantly inhibited by the tetracycline-inducible CRISPR/Cas9 system. Functional assays demonstrated that this system could inhibit proliferation, induce apoptosis and suppress cell migration. Therefore, the tetracycline-inducible CRISPR/Cas9 system was demonstrated to repress the malignant behavior of bladder cancer cells by controlling the expression of Cas9 and simultaneously targeting two oncogenic lncRNAs.
Collapse
Affiliation(s)
- Lu Peng
- Department of Clinical Laboratory, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Peng Pan
- Reproductive Medicine Center, Nanjing General Hospital, Nanjing, Jiangsu 210000, P.R. China
| | - Jinbu Chen
- Department of Clinical Laboratory, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Xueyuan Yu
- Department of Clinical Laboratory, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Jun Wu
- Department of Clinical Laboratory, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yong Chen
- Department of Clinical Laboratory, Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|