I. Introduction
Gene editing technologies have revolutionized the field of genetic medicine, offering unprecedented opportunities to correct disease-causing mutations at their source. Among these tools, CRISPR-Cas, Base Editors (BEs), and Prime Editors (PEs) stand out as transformative innovations. CRISPR-Cas9, derived from bacterial immune systems, laid the foundation for modern gene editing by enabling targeted DNA double-strand breaks (DSBs). Building on this, Base Editors and Prime Editors have emerged as advanced tools that offer single-nucleotide precision without inducing DSBs, reducing the risks associated with traditional CRISPR systems.
The therapeutic potential of these gene editing tools is immense, particularly for treating genetic disorders, cancers, and infectious diseases. However, their success hinges on the development of efficient delivery systems capable of safely and precisely delivering these tools to target cells. This article explores the mechanisms, innovations, and challenges of CRISPR, Base Editors, and Prime Editors in gene therapy, with a focus on delivery strategies, real-world applications, and the biotech companies leading the charge in this field.
II. CRISPR-Cas Systems
a. CRISPR-Cas9
The mechanism of the CRISPR-Cas9 system involves using a guide RNA (gRNA) to direct the Cas9 nuclease to a specific DNA sequence, where it introduces a DSB. The cell repairs the break through Non-Homologous End Joining (NHEJ), which often results in insertions or deletions (indels) leading to gene disruption, or Homology-Directed Repair (HDR), which allows precise gene correction using a donor template. This system is widely used for gene knockout, gene correction, gene insertion, and transcriptional regulation, offering high efficiency, versatility, and ease of use. However, it carries the risk of off-target effects and unintended mutations due to DSBs. Several companies are advancing CRISPR-Cas9-based therapies: CRISPR Therapeutics is developing CTX001 for sickle cell disease and beta-thalassemia, with promising clinical trial results; Editas Medicine is advancing EDIT-101 for Leber congenital amaurosis, a genetic blindness disorder; and Intellia Therapeutics is working on NTLA-2001 for transthyretin amyloidosis, which has shown positive Phase 1 clinical trial results.
b. CRISPR-Cas12 (Cpf1)
CRISPR-Cas12, similar to Cas9, creates staggered cuts (sticky ends) instead of blunt ends, which can improve HDR efficiency. This system is used for gene editing and diagnostics, such as the DETECTR platform for rapid viral detection. One of its key advantages is its smaller size compared to Cas9, which makes it easier to package into delivery vectors like adeno-associated virus (AAV), and it has shown reduced off-target effects in some cases. However, it is less widely adopted than Cas9, limiting its current applications. Mammoth Biosciences is a leading company in this space, developing CRISPR-Cas12-based diagnostics, including the DETECTR platform, which has been used for rapid and accurate detection of viral pathogens.
c. CRISPR-Cas13
CRISPR-Cas13 is a unique gene editing tool that targets RNA instead of DNA, making it particularly useful for RNA editing, viral RNA targeting, and diagnostics, such as the SHERLOCK platform for RNA detection. One of its key advantages is that it poses no risk of genomic DNA damage, as it operates exclusively at the RNA level. However, its applications are limited to RNA, which restricts its use compared to DNA-targeting systems like CRISPR-Cas9. Sherlock Biosciences is a prominent company leveraging CRISPR-Cas13 technology, developing the SHERLOCK diagnostics platform for rapid and accurate detection of RNA, including applications for COVID-19 and other infectious diseases.
d. CRISPR-CasΦ (Cas12f)
CRISPR-CasΦ (Cas12f) is an ultra-compact Cas protein, measuring less than half the size of Cas9, which allows it to be packaged into smaller delivery vectors like AAV. This makes it particularly useful for gene editing in space-constrained delivery systems, where larger Cas proteins may not fit. Its small size and efficient editing capabilities are significant advantages, though it is still in the early stages of development, limiting its current applications. Stella Therapeutics is one of the companies exploring the potential of CasΦ for gene therapy applications, with preclinical studies currently underway to evaluate its efficacy and safety.
III. Base Editors: Precision Without DSBs
Base Editors are engineered fusion proteins that combine a catalytically impaired Cas9 (nickase) with a deaminase enzyme, enabling precise single-nucleotide changes without inducing DSBs. There are two main types: Cytosine Base Editors (CBEs), which convert C•G to T•A, and Adenine Base Editors (ABEs), which convert A•T to G•C. These tools are particularly useful for correcting point mutations associated with genetic diseases, such as sickle cell anemia and progeria. A key advantage of Base Editors is their reduced risk of indels and off-target effects compared to traditional CRISPR-Cas9 systems. Several companies are actively working on Base Editor technologies, including Beam Therapeutics, which is developing BEAM-101 for sickle cell disease and BEAM-201 for cancer immunotherapy, and Verve Therapeutics, which is advancing VERVE-101 for familial hypercholesterolemia. These companies are making significant progress in preclinical and clinical studies, demonstrating the potential of Base Editors for precise and safe gene editing.
IV. Prime Editors: Versatile and Precise
Prime Editors utilize a Cas9 nickase fused to a reverse transcriptase and a prime editing guide RNA (pegRNA). The pegRNA directs the editor to the target DNA site and provides a template for precise DNA editing, enabling insertions, deletions, and base substitutions with high precision and minimal off-target effects. This system offers broader editing capabilities compared to Base Editors, making it suitable for correcting a wide range of genetic mutations, including those that are challenging to address with other gene editing tools. Companies like Prime Medicine are at the forefront of developing Prime Editor technologies, focusing on applications for genetic diseases such as Duchenne muscular dystrophy and cystic fibrosis.
V. Choosing the Right Delivery Vector
Selecting the appropriate delivery vector for gene editing tools depends on several critical factors. First, cargo size is a key consideration, as advanced editors like Prime Editors and Base Editors require larger delivery capacity compared to the more compact CRISPR-Cas9. This makes it challenging to package these editors into smaller vectors like AAV, often necessitating the use of dual-vector systems or alternative delivery methods. Second, target tissue specificity plays a significant role; for example, AAV vectors are ideal for tissue-specific delivery due to their natural tropism for certain organs, while lipid nanoparticles (LNPs) offer greater flexibility for systemic applications, such as targeting the liver or immune cells. Third, the safety profile of the delivery system is crucial, as minimizing immune responses and off-target effects is essential for clinical success. Finally, the choice between ex vivo vs. in vivo applications must be considered. Ex vivo editing, such as modifying CAR-T cells in the lab before reinfusion, allows for precise control over the editing process, while in vivo delivery requires robust and safe vectors that can efficiently deliver gene editing components to target tissues without causing adverse effects. Balancing these factors is essential to ensure the efficacy, safety, and success of gene editing therapies.
1. Anzalone, A. V., et al. (2019). “Search-and-replace genome editing without double-strand breaks or donor DNA.” Nature, 576(7785), 149-157.
2. Gaudelli, N. M., et al. (2017). “Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage.” Nature, 551(7681), 464-471.
3. Doudna, J. A., & Charpentier, E. (2014). “The new frontier of genome engineering with CRISPR-Cas9.” Science, 346(6213), 1258096.
4. Anzalone, A.V., et al. (2020). Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol, 38, 824–844.
PackGene Biotech is a world-leading CRO and CDMO, excelling in AAV vectors, mRNA, plasmid DNA, and lentiviral vector solutions. Our comprehensive offerings span from vector design and construction to AAV, lentivirus, and mRNA services. With a sharp focus on early-stage drug discovery, preclinical development, and cell and gene therapy trials, we deliver cost-effective, dependable, and scalable production solutions. Leveraging our groundbreaking π-alpha 293 AAV high-yield platform, we amplify AAV production by up to 10-fold, yielding up to 1e+17vg per batch to meet diverse commercial and clinical project needs. Moreover, our tailored mRNA and LNP products and services cater to every stage of drug and vaccine development, from research to GMP production, providing a seamless, end-to-end solution.
Related News
Nanoscope Therapeutics Publishes Breakthrough Clinical Data Demonstrating Significant Vision Restoration in Retinitis Pigmentosa with Novel AAV-based MCO-010 Gene Therapy
Nanoscope Therapeutics has announced the publication of highly encouraging Phase 1/2a clinical trial data in the esteemed journal Molecular Therapy, showcasing significant vision restoration in patients suffering from retinitis pigmentosa (RP), a debilitating...
Capsida’s AAV CAP-004 Gene Therapy Shows Promise for Friedreich’s Ataxia in Primate Study at MDA Conference
Capsida Biotherapeutics' experimental gene therapy, CAP-004, for Friedreich's ataxia (FA), has demonstrated promising preclinical results in nonhuman primates. Research presented by Capsida at the 2025 MDA Clinical & Scientific Conference showed that a single...
FDA Grants Fast Track to Sanofi’s Chlamydia mRNA Vaccine
Sanofi is making significant strides in the fight against chlamydia, the most common sexually transmitted bacterial infection, for which there are currently no approved preventative vaccines. The French pharmaceutical group has recently been granted a fast-track...
Epicrispr Biotechnologies Announces $68M Series B to Advance First-in-Class FSHD Epigenetic Therapy to Clinic
Epicrispr Biotechnologies today announced the first close of its Series B financing, securing $68 million to support the clinical development of EPI-321, a first-in-class, disease-modifying therapy for facioscapulohumeral muscular dystrophy (FSHD), a genetic...
Related Services

AAV CRISPR Plasmid
READ MORE

Lentivirus CRISPR Plasmid
READ MORE

Gene Editing mRNA
READ MORE