Journal of RNA and Genomics

Reach Us +44 1400530055

Review Article - Journal of RNA and Genomics (2021) Volume 17, Issue 3

Overview of Delivery of CRISPR/Cas Systems, Its Types and Role in Genome Editing and Immunotherapy

Awais Ali1*, Sadia Aslam2, Sonia Tabasum1, Rubab Aslam3

1Department of Biochemistry, University of Agriculture Faisalabad, Pakistan.

2Department of Biochemistry, Government College Women University Faisalabad, Pakistan.

3Department of Agriculture, University of Agriculture Faisalabad, Pakistan.

*Corresponding Author:
Awais A
Department of Biochemistry
University of Agriculture Faisalabad, Pakistan
E-mail: malikawaisali186@gmail.com

Received Date: 23 June 2020; Accepted Date: 22 July 2020; Published Date: 28 July 2020

© Copyright: Awais Ali, et al. First Published by Allied Academies. This is an open access article, published under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0). This license permits non-commercial use, distribution and reproduction of the article, provided the original work is appropriately acknowledged with correct citation details.

Visit for more related articles at Journal of RNA and Genomics

Abstract

A versatile programmable tool for effective and precise genome editing and gene targeting has been utilized and prepared with normal Clustered regularly interspaced short palindromic repeat-CRISPR-associated protein (CRISPR-Cas) genome editing, a prokaryotic adaptive immune system. This ground breaking approach can be used for the treatment, detection and treatment of cells, in vivo gene therapy or animal models. Safe and effective provision of CRISPR/Cas9 remains one of the biggest therapeutic problems in recent years. Though in-vivo and in-vitro genome editing have many advantages, their severe obstacles, such as small insertion sizes, high risk of carcinogenesis, and stimulation of the immune system are causing many problems for laboratory and clinical applications. The power of the latest cancer therapeutic approach has been shown by the genetically engineered immune cells using chimeric antigen receptors (CAR) or modified T cell receptors (TCR). While Autologous CD19 CAR T cells are effective in their clinical development, they have many advantages over their autologous counterparts, and recently, due to the advent of multiplex genome editing techniques, in particular CRISPR/Cas systems, have gathered wide-ranging attention.

Keywords

Cas9-sgRNA, Immunotherapy, CAR-T cells, Nanoparticles, Genome Editing.

Introduction

Recent years have seen researchers from all over the world draw interest with CRISPR-Genome Editing technologies mediated by Cas9, as a multi-purpose editing tool (Yuen et al. 2017, Mou et al. 2017, Zuo et al. 2017, Xiong et al. 2019). The CRISPRCas 9 method composed of a single-guide RNA (sgRNA) and the Cas9 DNA endonuclease, the prior directing latter toward specific DNA sequence to cut the double-strand DNA specifically sites (Jiang & Doudna 2017). Presently, genome editing mediated in CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) has been widely applied for the selective editing of single or bounteous gens using singleguide RNA in different cell types and species for specific site identities (Doudna & Charpentier 2014, Sander & Joung 2014, Slaymaker et al. 2016). CRISPR is made up of two critical modules, namely crRNA (CRISPR RNA) and tRNA (Tracr RNA) and Cas9 (Sapranauskas et al 2011, Barrangou et al. 2007), the single DNA endonuclease. In the field of genome editing, the emergence of synthetic SgRNA can direct to Cas9 can be single guided by sgRNA to produce a particular sites double-stranded break in DNA (DSBs) in queechy proto-spacer adjacent motif (PAM) targeted of genomic locus depending on a basis pair that can be mended by non-homologous end joining (NHEJ) or homologous direct repair (HDR) mechanism (Wang et al. 2013, Shalem et al. 2014, Cong et al. 2013, Moses et al. 2018).

Basically, two types of CRISPR systems were classified. here’s discussed were divided each in three types (class 1, Type I, III, IV and class 2, II, V and VI) (Barrangou & Horvath 2017, Makarova et al. 2017). CRISPR Type I, Type III both have used a many-cas protein complexes. CASCADE (CRISPRassociated antiviral defense complex) type I mechanism is capable of recognizing objective DNA (Zhang et al. 2012, Hochstrasser & Doudna 2015). Cas3 will subsequently disturb the destination loci. Cas10 is part of a multi- subunit complex like a cascade that recognizes and degrading invasive RNA in the Type III systems, a more general archaea System (Hsu et al. 2014). Meanwhile, types II only require protein from Cas9 to recognize, bind and degrade DNA (Brouns et al. 2008, Wu et al. 2015). The CRISPR could be transcribed into the precursor and then formed CRISPR RNA forms during the immune response called CRISPR RNA. The Cas nucleases, led by CRISPR RNA (crRNAs) can therefore accurately identify and split homology invaders nucleic acid (Wiedenheft et al. 2012). Progress in CRISPR/Cas device implementation has revolutionized field from fundamental to theoretical translation science.

The tripartite association of phage-Wolbachia-host is intriguing and yet remains unexplored. Role of WO phage in cytoplasmic incompatibility which itself is a tool of Wolbachia persistence inside the host is challenging and needs deeper insight. In the present work, we detected the presence of WO phage in five natural populations of Drosophila host and performed a comparative analysis of the Cif genes linked with the eukaryotic association model of phage.

Crispr/Cas Systems Toolbox

New developments in modulation mechanisms which originating from prokaryotes significantly support knowledge of the mechanism of tumor genesis. CRISPR-Cas is an adaptive immuno-responsive system that prevents infection by phages, viruses and other exotic genetics components in most bacteria and archaea (Deveau et al. 2010). Currently, CRISPR-Cas systems are categorized in two large classes. Class I structures form several subunit effectors complexes, where a wide variety of Cas proteins are linked to crRNA. We mainly discuss Class II system utilized as (Cas9, Cas12, and Cas13) a single nuclease such for genome editing which are used for delivery ejectives (Horvath & Barrangou 2010).

CAS 9

Three different methods allow CRISPR-mediated genome editing. The simplest and usually applied technique is Cas9– gRNA ribonucleoprotein (RNP) systems distribution to target cells (Makarova et al. 2011). A two in one plasmid (pX330, add gene plasmid #42230) of the Zhang Group was the primarily CRISPR-Cas9 of eukaryotic cells (Cong et al. 2013). Following the tradition of cancer care, surgical therapy shows that radiation therapy was the earliest treatment choice. Surgical and radiotherapy treatment is largely appropriate against localized non-metastatic cancers. The first Systemic cancer treatments were developed near 1940 through hormonal therapy, antimetabolite and chemical therapy for purpense treatment of non-metastatic, as well as metastatic cancer (Ran et al. 2013). The plasmid is exposed in two cassettes: one uses a U6 promoter to drive the expression of GRNA, the other uses a β-actin promoter for chicken to drive the expression of SpCas9. The double-strand genome diagnosis of DNA (Jinek et al. 2012) was allowed by Cas9, a crRNA-guided endonuclease consisting of HNH and RUVC nuclease domains

In order to extend the spectrum of target locus variants of the Cas9 Orth ologists with clearest protospacer adjacent motif (PAM) identifications capability was found. Staphylococcus aureus Cas9 (SaCas9) for instance will identify the 5′-NNGRRT sequence (Ran et al. 2015). During 2018, David Liu et al. (Hu et al. 2018) created Cas9 by phage-assisted continuous evolution, which recognizes multiple PAMs sequences (5′-NNGRRT, NNGAGT, etc.). Nishimasu and his colleagues formed SpCas9- NG in the latter half of the same year, which is able to identify relaxed NG PAM (Nishimasu et al. 2018). (Figure 1).

Figure

Figure 1: Cas9: The spectrum of target locus variants of the Cas9 orthologists with clearest protospacer adjacent motif (PAM) identifications capability was found. Cas9 by phage-assisted continuous evolution, which recognizes multiple PAMs sequences.

CRISPR CAS 12A

CRISPR/Cas9 research advancements provide encouragement to explore emerging systems for the development of applications in a comprehensive manner. Cas12a is one more DNA targeting method of CRISPR for specifically genome editing (called Cpf1, Figure 2). More specifically, Cas12a is an enzyme without tracrRNA assistance that is single crRNAguided and is responsible for precrRNA treatment. In this study we have reported the dual amplitude bio-sensing method based on CRISPR/Cas12a in order to successfully apply CRISPR/ Cas12a-based biosensor applications to biological enzymes (Ran et al. 2013). CRISPR/Cas12a will simplify the multiplex editing of the genome, hence its use may be much wider in tumor therapy in the future. The cleavage mechanism mediated by Cas12a is 0.126 μM EnGenLba, 1 NEB 2.12 buffers, crRNA (CRISPR RNA) 0.50 μM, ssDNA reporter 2.50 μM, RNase 10.0U inhibitor, 5.0 μL amplified products in a 20.0 μL reactions volumes. The reactions were performed averaged at 37°C for minimum 20 min on the Light-cycler 480 system with Fluorescence sampled every 30 seconds. The results were showed that the Cas12a cleavage system held on top of the capillaries after that thermocycling procedure due to the liquid surface tension within the capillary and air pressure in the limited cross-sectional area (Jinek et al. 2012). (Figure 2)

Figure

Figure 2: CRISPR-Cas12a (Cpf1) System. While Cas9 remains the best-characterized and most widely used nuclease for gene editing, Cas12a (previously named Cpf1) has recently emerged as an alternative. Cas12a has several unique features that distinguish it from Cas9 and expand the range of CRISPR-based genome editing tools. Most notable is the fact that Cas12a targets AT-rich regions of the genome, in contrast with Cas9, which targets GC-rich sequences.

CRISPR CAS 13A

In virus detection, Cas13a has already been applied successfully. However, it’s potential in treating cancer has not been fully explored as a new tool for gene interference. Recently found in bacteria, another innovative, rNA-guided, Cas13a (C2c2) enzyme originally known as C2c2, contains too superior, eukaryote and prokaryote nuclear-binding domains related to ribonucleases (Ran et al. 2015). Cas13a, initially known as the RNA target, will activate the following recognition and binding of the target RNA. The Cas13a can be used as a guided RNA, CRISPR-Cas RNA-target effector (Figure 3). CAS13a can be expressed heterologically in mammalian & plant cells and achieve knockdowns of correspondent or endogenous transcription with knockdown efficiency levels that are comparable to RNAi, but with considerably less off-target impact. The unique characteristic of CRISPR-Cas13a is that it is possible for Cas13a, CrRNA to be Enabled in isothermal condition by its target RNA to not specially cleave neighboring RNAs (Hu et al. 2018). A system using crRNA-controlled Cas 13a endo-nucleases for targeting RNA attracts considerable attention (Nishimasu et al. 2018). (Figure 3)

Figure

Figure 3: A CRISPR-Cas13 system. CRISPR-Cas13 is distinct and developed for targeting and editing of RNA from CRISPR-Cas9. A complex of CRISPR-Cas13a and CRNA ribonucleoprotein that is known as the target mRNA and causes sequence degradation. B REPAIR fuses catalytically damaged Cas13b (dCas13b) for editing single base mRNA adenosine deaminase (ADAR) without allowing RNA degradation to be converted into anosine. (REPAIR, programmable RNA editing A to I (G) substitution).

Delivery System

Presently, it remains a major problem for gene therapy how CRISPR-Cas components are efficiently delivered for precise genome editing to cells, tissues and organ. The benefits of the non-toxicity, well-targeted property, highly efficient, cheap price and low toxicity of good supply vectors should be provided (Lee & Kim 2019, Sahel et al. 2019). The CRISPR-Cas RNP or plasmid in-vivo genome version was placed by scientists from electroporation in cutaneous stem cells or mouse pancreas (Wu et al. 2017, Maresch et al. 2016). Protein expression in Cas9 can lead to target cells and ultimate editing of Cas9-mediated genes (Liang et al. 2015), as well as to the transmission of DNA or mRNA (plasmid or virus). Currently, in-vitro/ex-vivo and in-vivo CRISPR/Cas distribution can be split into two main groups, including an adjustable number of programmable node nucleases that reduce the probability of potential non-target genome editing and immunogenicity inductions of the immune response (Chen & Gonçalves 2016). In order to generate gene editing programs for cancer cells and immune system physical methods as well as for viral vectors and non-viral vectors, CRISPR/KS systems were used. CRISPR/Cas gene editing is an essential application in immunotherapy. For CAR-T therapy or allogeneic CAR-T cell growth CRISPR/Cas can be used. It can also be utilized in-vivo to modulate immune cells to regulate the expression of aberrant genesor to recognize and destroy cancerous cells of the body by immune cells (Zhang et al. 2019).

In-vivo CRISPR/Cas delivery

While in-vitro/ex-vivo approaches are the de facto norm for gene editing in applications for immunotherapy, the design of systems which modify cells in-vivo is advantageous. In-vivo procedures remove the need to remove or reintroduce cells into the patient as it is healthy and successful (Wan et al. 2019). It is important to explore riskless and efficient in vivo delivery systems in order to translationally incorporate CRISPR/ Cas systems in tumor therapy. In-vivo gene editing of tumor cells or the immune cells in both viral and non-viral vectors was developed in CRISPR/Cas systems (Kay 2011, Yin et al. 2014). Together, CRISPR-Cas9 therapeutic techniques are beneficial to facilitate the genetic modification of immune and cancer-cell cells quickly, including ex-vivo and in-vivo editing (Yin et al. 2016). CRISPR-Cas genetic disease circuits into CAR-T cells will also contribute in future attempts to improve programmability, to the effective, safer treatment of CRISPR and thereby to cell-based, programmable and intelligent medicines for cancer treatment. CRISPR tests were used to detect genes involved in a wide range of processes, including drug-resistant regulators (Hou et al 2017) synergy and synthetic lethal associations (Konermann et al. 2015) PD-L1 expression regulatory authorities and core genes (Hart et al. 2015).

Viral Vector: CRISPR-Cas9 viral systems for transmission, Like lentivirus, adenovirus and adeno-associated virus (AAV) (Yin et al. 2017). These include, in particular, the most sophisticated in vivo gene-supply technique (Ran et al 2015, Yang et al. 2017), examples of active treatment of mouse models of neuro-degenerative diseases (Gaj rt al. 2017) and of their efficacy and protection in clinical trials evaluated and recently approved, other CRISPR-Cas viral vectors remain worthy of further in vivo exploration. For CRISPR/Cas systems in tumor treatment, viral vectors are considered to be successful. Liu and his co-worker have used CRISPR/Cas9, a method based on lentivirus, to treat the human HCC using the sgRNA-721 (LV-H721) HIF-1a (Liu et al. 2018). In the subcutaneous HCC model of SMMC-7721, they inserted LV-H721 directly into the tumor tissues. The CRISPR/Cas9 lentivirus-mediated tumor tissue decreased significantly 3 days after therapy the expression of HIF-1α. Lentivirus and AAV expressed in SW403 cells, Cas9 and the SgRNA, used by Kim and coworkers to attack mutant KRAS in alone (~50%) (Kim et al. 2018). The tumor development has been successfully blocked with intra-tumor injection in the subcutaneous xenograft colon carcinoma model.

Non-viral vector: The different stages of clinical development for RNA-based therapeutics are non-viral approaches like nanoparticles of lipid or polypeptide. In comparison with AAV, the CRISPR-Cas component nano particle-based delivery has a high nuclear acid cargo loading capacity, without the possibility of genomic incorporation, and without the results from persistent CRISPR-Cas9 expression (Kaczmarek et al. 2017).

Common nano-sized preparations: Non-viral techniques such as lipid and polypeptide nanoparticles form part of the various stages of clinical research of RNA-based therapy. In contrast to AAVs, components of CRISPR-Cas have a high loading capacity for nuclear acid cargoes without genomic integration and/or persistent CRISPR-Cas9 expression (Kaczmarek et Al. 2017) in nanoparticles. The complexes of Cas9-sgRNA RNP will effectively be transported with cationic lipids to the inner octrois of the mouse (Zuris et al. 2015), thus increasing the degree of hearing loss (Gao et al. 2018), allowing possible future use for skin cancers such as melanoma. For example, the mouse brain that was mediated by the provision of engineered Cas9 RNP complexes with many sequences of nuclear location in SV40 was recently identified in active neuronal surveying over these last years. Case CRISPR Cas9/sgRNA plasmid 1 (Plk1) can also be used to use polythene phospholipid-modified cationic nanoparticle’s, which are modified by the polythene cationic lipid nanoparticles (PLNP). A strongly preserved serine-threonine kinase which promotes cell division and overexpresses it in different tumors (Staahl et al. 2017. The Plk1 protein has been decreased dramatically in the form of i.t. injection in subcutaneous melanoma mice derived from A375, and in vivo development (N67 percent) has been inhibited. Increasing programming can also continue to enhance and increase the safety of CRISPR treatment in future, with a view to developing a CRISPR-based gene disrupting circuit into CAR-T cells in the future, programmable and intelligent cell medicine for cancer care (Eckerdt et al. 2005).

In-Vitro delivery system of CRISPR/Cas

It is successful to obtain optimum CRISPR/Cas tools depend on the in-vitro trials with high activity of gene editing for cancer treatment (Zhang et al. 2017). The power of advanced genetics and reverse genetics is combined in high performance screens, especially across the genome. CRISPR/Cas is now operating in-vitro or ex-vivo in all therapeutic applications. At present, CRISPR/Cas immunotherapy uses have been restricted mainly to the expansion of multiple immunes cells, T cells including B cells, dendrite cells and NK cells (Yin et al. 2018, Lenz et al. 2003, Hung et al. 2018, Kararoudi et al. 2018). Several strategies of Non-viral and viral transmission are discussed.

Non-viral: Researchers also have recently been able to supply cells directly with the Cas protein or RNP (Hung et al. 2018). Electroporation is now the most effective physical means of gene distribution. An electric field is applied to the target cells, small pores in the cell membrane are temporarily opened to allow DNA or other molecules to enter the cell (Rols 2017). Electroporation is also favored because it normally contributes to high efficiencies in transfection. The provision of massive gene editing methods such as CRISPR/Cas systems using conventional cationic transfection reagent’s continues to be difficult. Su and his colleagues have shown that the supply of encoding sgRNA plasmid and Cas9 into primary T cells of human by electroporation has been able to efficiently eliminate PD-1 expression (Rols et al. 2016). Researchers have shown the viability of the primary human T cells in extend in-vitro culture, while the PD-1 gene expression has been decreased. The cells displayed an increase in the synthesis and increased cytotoxicity of the up-regulated interferon-μ (IFN-α). Direct modulation of human immune cells may be synergistic with current T-cell therapies. Xing and colleagues invent a nano carry of a new CRISPR/Cas9 complex on the basis of GO (graphene oxide) – PEG (polyethylene glycol) – PEI (polyethyleneimine). AGS cell expressed the GFP via endocytosis and endosome escape, reaching therefore 39% of a GFP gene editing efficiency, will successfully join CRISPR/Cas9 in the nanocarrier. The efficiency of in -vivo transmission in most of the above-noted new carriers is worth investigating, according to the high efficiency with regard to in vitro gene editing (Yue et al. 2018).

Viral delivery: The viral vectors are one of the famous CRISPR/Cas granting methodologies (Xu et al. 2019). Different viral transfer substructure has been utilized for transportation of CRISPR/Cas Substructure into tumor cells in vitro. AAV has been to a great extent applied for CRISPR genome altering because of its great security displayed in numerous clinical preliminaries. But a single AAV carrier size (4.7kb) limits the device kit of the CRISPR/Cas encyclopedia of large genes. While in-vitro transformation to cells can promptly be accomplished through electroporation, among other actual techniques, a more secure and more useful skeleton is needed for in vivo applications in human quality treatment. Adenovirus, a non-inclusive, double abandoned DNA (dsDNA) icosahedral capsid infection, is suited for concentrated transport by collaboration with innumerable human cell surface receptors. This large tropism and simplicity of change make the CRISPR-Cas9 substructure of this form of infection attractive for quality transmission (Xu et al. 2019). The development of lentiviral depends largely on the transfection of four plasmids into HEK293T cells (three bundles develop and a vector of exchange) (Ryø et al. 2019). CRISPR/cas Structure for the use of lentiviral vectors subsequently becomes inefficient and cumbersome in vitro.

Anti-Tumor Targets for Gene Editing

It is basic for effective CRISPR-based antitumor treatment to choose a fitting quality objective to amplify adequacy just as limit harmfulness. The thought of restorative focuses for malignant growth treatment includes expound connections among tumor, host and climate which will impact the treatment impact of CRISPR/Cas-based frameworks (Lee & Kim 2019, Sahel et al. 2019, Wu et al. 2017).

Immunogenicity

CRISPR-Cas system parts are derived from microorganisms, Cas content reactions are unsusceptible, and Cas protein can be seen as the major difficulties for the CRISPR-Cas clinical preparations (Zou 2005, Gajewski et al. 2006, Zitvogel et al. 2006). The immunogenic cell passing that can get the versatile safe reaction can likewise be utilized as the readout. For instance, Shi et al. utilized the cell suitability as the read out to examine the critical atoms in pyro ptosis, an incendiary type of customized cell passing, which instigates immunogenic cell demise (Dunn et al. 2002). Immune avoidance is fundamental for disease development and movement. This might be likewise the reason for tumor opposition against traditional immunotherapy. (Zou 2005, Gajewski et al. 2006, Zitvogel et al. 2006, Dunn et al. 2002). In order to elevate healthy cell properties, the CRISPR/ Cas9 techniques were used. The CAR-T cells were engineered to increase tumor limit enemies in the tumor-bearing model with the disturbance of the personalized cell passing 1 (PD-1) (Ren et al. 2017). CD47 disorder demonstrated an anticipated immunotherapy treatment for small cells in the lungs. (Weiskopf et al. 2016).

Clinical Translation

In 2016 the West China Hospital, Sichuan University, launched the first clinical trial for CRISPR/Cas in the treat of pulmonary cancer. All of the CRISPRCas9 technology clinical experiments, regardless of the status or form of illness (Kosicki et al. 2018). The PD-1 immune control point receptor was first knocked out ex vivo in the blood T cells of the recipient with the help of CRISPR/ Cas system, in a non-randomized, open label Phase I analysis of T-cells in lung cancer patients (NCT0202793856), oesophageal cancer patients (ClinicalTrials.gov: NCT03081715). In order to evaluate security after normal therapy in the treatment of metastatic NSCLC, the PD-1, knockout engineered T cells have been expanded and then injected back into the patients. Related PD-1 T-cells designed for knockout were registered in four clinical tests respectively, to treat bladder cancer, breast, renal and oesophageal cancer (Scott & Zhang 2017). For instances, research is underway into the effects of mesothelin-positive solid tumor’s (Clinical Trials. gov: NCT03545815) of PD1- and TCR-knockout anti-mesothelin CAR-T cells controlled by CRISPR-Cas9.Additional clinical trials have been started by the general hospital of the Baylor College of Medicine and the Chinese People’s Liberation Army (PLA) to investigate the reaction of T-cell and B-cells to CRISPR-Cas9-modified CAR-T cells (Clinical Trials.gov: NCT03690011, NCT03398967, and NCT03166878). They knock out genes to increase anti-tumor reactivity, tumor specificities, or tumor immune suppressive effects on the surface of CAR-T cells (Haapaniemi et al. 2018).

In Ex-vivo methods, the manipulated cells can be controlled more efficiently than in-vivo application and the desired clone selected (Schumann et al. 2015). Due to the wide GMP ex-vivo and the high accessibility of hématophytic cells, the majority of ex-vivo applications concentrate on those cells. Now they use electronics and viral vectors to achieve surprisingly high levels of gene editing efficiencies on T cells ex-vivo. The NHEJ mediated gene disorders usually achieve efficiency indels of more than 80% while the editing of HDR mediated genome leads to changes in primary human T cells of around 30 to 70% (Porteus 2019). Hereditary illnesses (for instance β-Thalassemia, sickle-cell anemia...), viral inhalation and cancer immunotherapy are three major applications of ex-vivo gene editation. CRSIPR/Cas9 is used locally or systemically for in-vivo use of contaminated cells or organs in the body. Invivo. Monogenic genetic defects are the main targets of in-vivo therapy (e.g., Duchen muscular dystrophy (Min et al. 2019), tyrosinemia etc.). Since NHEJ is higher than HDR, the main application is the NHEJ pathways, which contribute to gene disruption. The second uses the AAV service retinal cavity (Yu et al. 2017). Yu el al. Cas9 induced gene disorders preserved the function of a cone photoreceptor in three different models of retinal degeneration mouse. The first in-vivo clinical trial was recently launched on the basis of a corresponding technique for Leber Congenital Amaurosis 10 (LCA10) (Maeder et al. 2019).

Concluding Remarks

The progression in the development of CRISPR systems has brought advantages to the clinical application of cancer, in view of the brief history of CRISPR technology. CRISPR/Cas9 has been used extensively in human genomic DNA, recently aimed to target viral diarrhea in ho, and it has been implemented to date to enable genome-focused, loss-of-function or gain of function (CRISPRa), in-vitro, ex-vivo and/or in-vivo trials that have not been completed in all the existing cancer therapeutic strategies. This review summarizes the application of CRISPR/Cas in abolishing these viruses from the host by directly targeting viral genomes or particular host genes that aid in viral replication and persistence.

In recent years, we have witnessed an incredible increase and expansion in cancer immunotherapy, particularly in the use and interface with the rapid development of genome editing techniques of genetically modified immune cells. Recent technical capacity for the treatment of solid cancer tumors with the use of T-cells has shown significant progress in preclinical studies, but still faces many challenges that prevent the success of clinical trials. The advancement of adoptive cell therapy can only be made a consistent, safe, and effective platform in cancer care with an exact combination of effectiveness and toxicity.

References

  1. Abudayyeh OO, Gootenberg JS, Essletzbichler P, et al. 2017. RNA targeting with CRISPR–Cas13. Nature, 550(7675), 280-284.
  2. Abudayyeh OO, Gootenberg JS, Konermann S, et al. 2016. C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science, 353(6299).
  3. Barrangou R & Horvath P. 2017. A decade of discovery: CRISPR functions and applications. Nat microbiol, 2(7), 1-9.
  4. Barrangou R, Fremaux C, Deveau H, et al. 2007. CRISPR provides acquired resistance against viruses in prokaryotes. Science, 315(5819), 1709-1712.
  5. Brouns SJ, Jore MM, Lundgren M, et al. 2008. Small CRISPR RNAs guide antiviral defense in prokaryotes. Science, 321(5891), 960-964.
  6. Chen JS, Ma E, Harrington LB, et al. 2018. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science, 360(6387), 436-439.
  7. Chen X, & Gonçalves MA. 2016. Engineered viruses as genome editing devices. Mol Ther, 24(3), 447-457.
  8. Chew WL, Tabebordbar M, Cheng JK, et al. 2016. A multifunctional AAV–CRISPR–Cas9 and its host response. Nat methods, 13(10), 868-874.
  9. Cong L, Ran FA, Cox D, et al. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science, 339(6121), 819-823.
  10. Deveau H, Garneau JE, & Moineau S. 2010. CRISPR/Cas system and its role in phage-bacteria interactions. Annu rev  microbiol, 64, 475-493.
  11. Doudna JA & Charpentier E. 2014. The new frontier of genome engineering with CRISPR-Cas9. Science, 346(6213).
  12. Dunn GP, Bruce AT, Ikeda H, et al. 2002. Cancer immunoediting: from immunosurveillance to tumor escape. Nat immunol, 3(11), 991-998.
  13. Eckerdt F, Yuan J, & Strebhardt K. 2005. Polo-like kinases and oncogenesis. Oncogene, 24(2), 267-276.
  14. Gaj T, Ojala DS, Ekman FK, et al. 2017. In vivo genome editing improves motor function and extends survival in a mouse model of ALS. Science advances, 3(12), eaar3952.
  15. Gajewski TF, Meng Y, Blank C, et al. 2006. Immune resistance orchestrated by the tumor microenvironment. Immunol rev, 213(1), 131-145.
  16. Gao X, Tao Y, Lamas V, et al. 2018. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature, 553(7687), 217-221.
  17. Haapaniemi E, Botla S, Persson J, Schmierer B, et al. 2018. CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response. Nat med, 24(7), 927-930.
  18. Hart T, Chandrashekhar M, Aregger M, et al. 2015. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell, 163(6), 1515-1526.
  19. Hochstrasser ML, & Doudna JA. 2015. Cutting it close: CRISPR-associated endoribonuclease structure and function. Trends biochem sci, 40(1), 58-66.
  20. Horvath P, & Barrangou R. 2010. CRISPR/Cas, the immune system of bacteria and archaea. Science, 327(5962), 167-170.
  21. Hou P, Wu C, Wang Y, et al. 2017. A genome-wide CRISPR screen identifies genes critical for resistance to FLT3 inhibitor AC220. Canc res, 77(16), 4402-4413.
  22. Hsu PD, Lander ES, & Zhang F. 2014. Development and applications of CRISPR-Cas9 for genome engineering. Cell, 157(6), 1262-1278.
  23. Hu JH, Miller SM, Geurts MH, et al. 2018. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature,556, 57–63.
  24. Hung KL, Meitlis I, Hale M, et al. 2018. Engineering protein-secreting plasma cells by homology-directed repair in primary human B cells. Mol Ther, 26(2), 456-467.
  25. Jiang F & Doudna JA. 2017. CRISPR–Cas9 structures and mechanisms. Annu rev biophys, 46, 505-529.
  26. Jinek M, Chylinski K, Fonfara I, et al. 2012. A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. science, 337(6096), 816-821.
  27. Kaczmarek JC, Kowalski PS, & Anderson DG. 2017. Advances in the delivery of RNA therapeutics: from concept to clinical reality. Genome med, 9(1), 1-16.
  28. Kararoudi MN, Dolatshad H, Trikha P, et al. 2018. Generation of knock-out primary and expanded human NK cells using Cas9 ribonucleoproteins. JoVE (J Vis Exp), (136), e58237.
  29. Kay MA. 2011. State-of-the-art gene-based therapies: the road ahead. Nat Rev Genet, 12(5), 316-328.
  30. Kim W, Lee S, Kim HS, et al. 2018. Targeting mutant KRAS with CRISPR-Cas9 controls tumor growth. Genome research, 28(3), 374-382.
  31. Konermann S, Brigham MD, Trevino AE, et al. 2015. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature, 517(7536), 583-588.
  32. Kosicki M, Tomberg K, & Bradley A. 2018. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat biotechnol, 36(8), 765-771.
  33. Lee M, & Kim H. 2019. Therapeutic application of the CRISPR system: current issues and new prospects. Hum genet, 138(6), 563-590.
  34. Lenz P, Bacot SM, Frazier-Jessen MR, et al. (2003). Nucleoporation of dendritic cells: efficient gene transfer by electroporation into human monocyte-derived dendritic cells. FEBS letters, 538(1-3), 149-154.
  35. Liang X, Potter J, Kumar S, et al. 2015. Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J biotechnol, 208, 44-53.
  36. Liu Q, Fan D, Adah D, et al. 2018. CRISPR/Cas9‑mediated hypoxia inducible factor‑1α knockout enhances the antitumor effect of transarterial embolization in hepatocellular carcinoma. Oncology reports, 40(5), 2547-2557.
  37. Maeder ML, Stefanidakis M, Wilson CJ, et al. 2019. Development of a gene-editing approach to restore vision loss in Leber congenital amaurosis type 10. Nat med, 25(2), 229-233.
  38. Makarova KS, Haft DH, Barrangou R, et al. 2011. Evolution and classification of the CRISPR–Cas systems. Nat Rev Microbiol, 9(6), 467-477.
  39. Makarova KS, Zhang F, & Koonin EV. 2017. SnapShot: class 2 CRISPR-Cas systems. Cell, 168(1), 328-328.
  40. Maresch R, Mueller S, Veltkamp C, et al. 2016. Multiplexed pancreatic genome engineering and cancer induction by transfection-based CRISPR/Cas9 delivery in mice. Nat commun, 7(1), 1-13.
  41. Min YL, Li H, Rodriguez-Caycedo C, et al. 2019. CRISPR-Cas9 corrects Duchenne muscular dystrophy exon 44 deletion mutations in mice and human cells. Sci adv, 5(3), eaav4324.
  42. Moses C, Garcia-Bloj B, Harvey AR, et al. 2018. Hallmarks of cancer: The CRISPR generation. European J Canc, 93, 10-18.
  43. Mou H, Smith JL, Peng L, et al. 2017. CRISPR/Cas9-mediated genome editing induces exon skipping by alternative splicing or exon deletion. Genome biol, 18(1), 1-8.
  44. Nishimasu H, Shi X, Ishiguro S, et al. 2018. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science, 361(6408), 1259-1262.
  45. Porteus MH. (2019). A new class of medicines through DNA editing. New England Journal of Medicine, 380(10), 947-959.
  46. Ran FA, Cong L, Yan WX, et al. 2015. In vivo genome editing using Staphylococcus aureus Cas9. Nature, 520(7546), 186-191.
  47. Ran FA, Hsu PD, Wright J, et al. 2013. Genome engineering using the CRISPR-Cas9 system. Nat protoc, 8(11), 2281-2308.
  48. Ren J, Liu X, Fang C, et al. 2017. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin cancer res, 23(9), 2255-2266.
  49. Rols MP. 2017.Parameters affecting cell viability following electroporation in vitro, Handb. Electroporation, Springer International Publishing, pp,1449–1465.
  50. Ryø LB, Thomsen EA, & Mikkelsen JG. 2019. Production and validation of lentiviral vectors for CRISPR/Cas9 delivery. In CRISPR Gene Editing (pp. 93-109). Humana Press, New York, NY.
  51. Sahel DK, Mittal A, & Chitkara D. 2019. CRISPR/Cas system for genome editing: progress and prospects as a therapeutic tool. J Pharmacol Exp Ther, 370(3), 725-735.
  52. Sahel DK, Mittal A, & Chitkara D. 2019. CRISPR/Cas system for genome editing: progress and prospects as a therapeutic tool. J Pharmacol Exp Ther, 370(3), 725-735.
  53. Sander JD & Joung JK. 2014. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat biotechnology, 32(4), 347-355.
  54. Sapranauskas R, Gasiunas G, Fremaux C, et al. 2011. The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. Nucleic acids res, 39(21), 9275-9282.
  55. Schumann K, Lin S, Boyer E, et al. 2015. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proceedings of the National Academy of Sciences, 112(33), 10437-10442.
  56. Scott DA, & Zhang F. 2017. Implications of human genetic variation in CRISPR-based therapeutic genome editing. Nat med, 23(9), 1095.
  57. Shalem O, Sanjana NE, Hartenian E, et al. 2014. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science, 343(6166), 84-87.
  58. Shi J, Zhao Y, Wang K, et al. 2015. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature, 526(7575), 660-665.
  59. Slaymaker IM, Gao L, Zetsche B, et al. 2016. Rationally engineered Cas9 nucleases with improved specificity. Science, 351(6268), 84-88.
  60. Staahl BT, Benekareddy M, Coulon-Bainier C, et al. 2017. Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nature biotechnology, 35(5), 431-434.
  61. Su S, Hu B, Shao J, et al. 2016. CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients. Sci rep, 6(1), 1-14.
  62. Wan T, Niu D, Wu C, et al. 2019. Material solutions for delivery of CRISPR/Cas-based genome editing tools: Current status and future outlook. Materials Today, 26, 40-66.
  63. Wang DX, Wang J, Du YC, et al. 2020. CRISPR/Cas12a-based dual amplified biosensing system for sensitive and rapid detection of polynucleotide kinase/phosphatase. Biosensors and Bioelectronics, 168, 112556.
  64. Wang H, Yang H, Shivalila CS, et al. 2013. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. cell, 153(4), 910-918.
  65. Weiskopf K, Jahchan NS, Schnorr PJ, et al. 2016. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer.  J clin invest, 126(7), 2610-2620.
  66. Wiedenheft B, Sternberg SH, & Doudna JA. 2012. RNA-guided genetic silencing systems in bacteria and archaea. Nature, 482(7385), 331-338.
  67. Wu W, Lu Z, Li F, et al. 2017. Efficient in vivo gene editing using ribonucleoproteins in skin stem cells of recessive dystrophic epidermolysis bullosa mouse model. Proceedings of the Natl Acad Sci, 114(7), 1660-1665.
  68. Wu Y, Zhou H, Fan X, et al. 2015. Correction of a genetic disease by CRISPR-Cas9-mediated gene editing in mouse spermatogonial stem cells. Cell res, 25(1), 67-79.
  69. Xiong X, Chen M, Lim WA, et al. 2019. CRISPR/Cas9 for human genome M. Chen et al. Cancer Letters ,447, 48–55.
  70. Xu CL, Ruan MZ, Mahajan VB, et al. 2019. Viral delivery systems for CRISPR. Viruses, 11(1), 28.
  71. Yang S, Chang R, Yang H, et al. 2017. CRISPR/Cas9-mediated gene editing ameliorates neurotoxicity in mouse model of Huntington’s disease. The J clin invest, 127(7), 2719-2724.
  72. Yin H, Kanasty RL, Eltoukhy AA, et al. 2014. Non-viral vectors for gene-based therapy. Nat Rev Genet, 15(8), 541-555.
  73. Yin H, Kauffman KJ, & Anderson DG. 2017. Delivery technologies for genome editing. Nat rev Drug discov, 16(6), 387.
  74. Yin H, Song CQ, Dorkin JR, et al. 2016. Therapeutic genome editing by combined viral and non-viral delivery of CRISPR system components in vivo. Nat biotechnol, 34(3), 328-333.
  75. Yin H, Song CQ, Suresh S, et al. 2018. Partial DNA-guided Cas9 enables genome editing with reduced off-target activity. Nat chem biol, 14(3), 311.
  76. Yu W, Mookherjee S, Chaitankar V, et al. 2017. Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice. Nat commun, 8(1), 1-15.
  77. Yue H, Zhou X, Cheng M, et al. 2018. Graphene oxide-mediated Cas9/sgRNA delivery for efficient genome editing. Nanoscale, 10(3), 1063-1071.
  78. Yuen G, Khan FJ, Gao S, et al. 2017. CRISPR/Cas9-mediated gene knockout is insensitive to target copy number but is dependent on guide RNA potency and Cas9/sgRNA threshold expression level. Nucleic acids res, 45(20), 12039-12053.
  79. Zetsche B, Gootenberg JS, Abudayyeh OO, et al. 2015. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell, 163(3), 759-771.
  80. Zhang J, Rouillon C, Kerou M, et al. 2012. Structure and mechanism of the CMR complex for CRISPR-mediated antiviral immunity. Mol cell, 45(3), 303-313.
  81. Zhang L, Wang P, Feng Q, et al. 2017. Lipid nanoparticle-mediated efficient delivery of CRISPR/Cas9 for tumor therapy. NPG Asia Materials, 9(10), e441-e441.
  82. Zhang Y, Shen S, Zhao G, et al. 2019. In situ repurposing of dendritic cells with CRISPR/Cas9-based nanomedicine to induce transplant tolerance. Biomaterials, 217, 119302.
  83. Zitvogel L, Tesniere A, & Kroemer G. 2006. Cancer despite immunosurveillance: immunoselection and immune subversion. Nat Rev Immunol, 6(10), 715-727.
  84. Zou W. 2005. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat rev cancer, 5(4), 263-274.
  85. Zuo E, Cai YJ, Li K, et al. 2017. One-step generation of complete gene knockout mice and monkeys by CRISPR/Cas9-mediated gene editing with multiple sgRNAs. Cell res, 27(7), 933-945.
  86. Zuris JA, Thompson DB, ShuY, et al. 2015. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat biotechnol, 33(1), 73-80.
Get the App