CBpromise   

Our promise to you:
Guaranteed product quality, expert customer support.

24x7 CUSTOMER SERVICE
CONTACT US TO ORDER

Solution for Vaccine Development Using CRISPR Technology    

In recent years, viral infections have posed significant threats to both human and animal health globally. For instance, highly pathogenic avian influenza viruses (AIVs) can potentially cause widespread infections and high mortality rates. The outbreak of the COVID-19 pandemic has further intensified the urgent need for vaccines and antiviral treatments. Vaccination is widely considered one of the most effective methods for preventing infectious diseases. Historically, global vaccination initiatives have eradicated deadly diseases such as smallpox and rinderpest. However, existing vaccine development technologies face various limitations, such as complex production processes, high costs, and the inability to rapidly respond to emerging viruses.

Development in Vaccine Technologies

The development of viral vaccines has undergone significant transformations, evolving from traditional inactivated or attenuated vaccines to recombinant vaccines using modern genetic engineering techniques. Live-attenuated vaccines have long been considered one of the best forms of vaccines due to their ability to elicit strong immune responses. However, producing live-attenuated vaccines is highly complex and costly. This process typically requires prolonged cultivation and extensive screening steps to ensure that the virus can effectively trigger an immune response without harming the host.

Advantages of CRISPR in Vaccine Development

The advent of CRISPR/Cas9 gene-editing technology offers new hope for the development of vaccines and antiviral therapies. This technology is advantageous due to its simplicity and low cost, making it widely applicable in the biomedical field.

Advantage 1: Multi-Gene Targeting Capability

The CRISPR/Cas system can simultaneously target multiple genes. By designing multiple sgRNAs, Cas9 can knock out or activate several genes at the same time. This capability allows researchers to comprehensively modify and evaluate complex viral genomes, significantly enhancing research efficiency.

Advantage 2: Studying Viral Evolution

CRISPR technology can be used to study viral evolution and predict potential future mutations. By analyzing viral genomes, scientists can obtain critical data that serves as a key reference for vaccine design.

Advantage 3: Integration with Other Technologies

The CRISPR/Cas system can be integrated with other modern biotechnologies, such as high-throughput screening and single-cell sequencing, to drastically accelerate the speed and efficiency of vaccine development. For example, in the development of COVID-19 vaccines, researchers used CRISPR to rapidly screen effective vaccine candidates and proceeded to mass production.

Advantage 4: RNA-Targeted Editing

While most CRISPR/Cas9 editing targets DNA viruses, the use of RNA-targeting CRISPR enzymes like Cas13 has become possible. This method applies to single-stranded RNA viruses (ssRNA) and double-stranded RNA viruses (dsRNA). For instance, Cas13d and specific crRNAs can target the RNA of SARS-CoV-2 and H1N1 viruses, effectively inhibiting viral replication.

FFigure. 1 illustrates the Application of CRISPR/Cas Systems for Antiviral Therapy, highlighting the distinct mechanisms of action against DNA viruses and RNA viruses. (doi: 10.1016/j.virusres.2024.199314)Figure 1. Utilization of CRISPR/Cas Systems in DNA Viruses and RNA Viruses. (Zahedipour F, et al., 2024)

Steps in Vaccine Development

  • Selection of Target Genes and Design of sgRNAs

First, it is necessary to identify the target genes for editing in vaccine development or antiviral therapy. For attenuated live-vaccine development, the target genes might be those related to viral virulence and pathogenesis; for CRISPR-based antiviral treatment, the target genes could be essential for viral replication or host infection. Using bioinformatics tools and experimental validation, researchers design specific sgRNAs to guide Cas9 in cutting the target genes.

  • Construction of the CRISPR/Cas Editing System

Once the sgRNAs are designed, the next step is constructing the appropriate CRISPR/Cas editing system. Typically, these vectors can be delivered into host cells via viral vectors (such as adeno-associated viruses, AAV) or non-viral vectors (such as plasmids or electroporation).

  • Validation in Cell and Animal Models

Before proceeding with vaccine development, the efficacy and safety of the CRISPR/Cas9 system must be validated in cell and animal models. For instance, gene knockout or activation experiments can be conducted on cell lines to detect the expression levels and functions of the respective gene products. Subsequently, edited viruses or cells are injected into animal models to evaluate immune responses and protective effects.

Comprehensive CRISPR Solutions for Advanced Vaccine Development

The application prospects of CRISPR technology in vaccine development are extensive. Using CRISPR/Cas9 technology enables large-scale evaluation and detection of genomic recombination points, addressing consistency issues in vaccines produced by traditional methods. Moreover, employing multiple CRISPR/Cas9 methods for gene editing can further enhance the immunogenicity of the virus and the efficacy of the vaccine. Despite numerous advantages, continued research and optimization are necessary to overcome challenges such as off-target effects, PAM limitations, and delivery difficulties, thereby achieving broader applications.

At Creative Biogene, we offer comprehensive technical support and product supply for vaccine development, leveraging advanced techniques such as CRISPR/Cas9 gene editing and RNA targeting. Our solutions enable simultaneous multi-gene targeting, virus genome modification, and high-throughput screening, accelerating vaccine design and production to address emerging viral threats efficiently.

Related Services and Products

CRISPR/Cas13 RNA Editing Services

CRISPR Library Screening Service

Gene Editing Cell Line Generation Service

AAV Service

Residual DNA Sample Prep Kit for Vaccine

References:

  1. Bhujbal S, Bhujbal R, Giram P. An overview: CRISPR/Cas-based gene editing for viral vaccine development. Expert Rev Vaccines. 2022 Nov;21(11):1581-1593.
  2. Zahedipour F, Zahedipour F, Zamani P, Jaafari MR, Sahebkar A. Harnessing CRISPR technology for viral therapeutics and vaccines: from preclinical studies to clinical applications. Virus Res. 2024 Mar;341:199314.
For research use only. Not intended for any clinical use.
Inquiry