Learn more Webcast featuring eSpOT-ON
Explore More eSpOT-ON Nuclease Protein Available Now
Explore More Order eSpOT-ON Nuclease mRNA Now
APPLICATIONS

CRISPR Base Editing

Adenine and Cytosine Base Editors Can Transform Therapeutic Gene Editing

Accubase™ Cytosine Base Editor

Available in Research-grade and GMP, RNP AccuBase base editor delivers high efficiency and exceptional fidelity, ensuring precise single base modifications with minimal off-target activity. Designed to meet stringent therapeutic development standards, AccuBase is a safe and reliable solution for advancing clinical and commercial applications.

Accubase™ Cytosine Base Editor

Your base editing success starts with Synthego, where advanced engineering and precision converge in our Accubase™ Cytosine Base Editor (CBE). Offering high efficiency and exceptional fidelity, AccuBase is engineered to meet the demanding requirements of therapeutic development. Available in both Research-grade and GMP, you have the flexibility to integrate AccuBase into your projects at any stage. By pairing AccuBase’s high efficiency base editing performance with Synthego’s best-in-class gRNAs, you can confidently tackle the complexities of single or multiplex base editing with solutions optimized for safety and consistency.

A significant number of human genetic diseases are caused by single nucleotide variants, mutations that involve the alteration of a single DNA nucleotide. In fact, an estimate of up to 90% of known pathogenic genetic variants are caused by SNVs. These mutations can disrupt essential biological processes and are implicated in a wide range of conditions, from inherited cancers to rare monogenic disorders. Also, data from the NIH’s All of Us Reseach Program unveiled over 275 million previously undocumented genetic variants, including nearly 4 million potentially disease relevant regions. This growing understanding underscores the critical need for precision gene editing therapeutics, using tools like CRISPR to correct the mutations that cause many of these genetic diseases.

CRISPR base editors are one type of gene editing tool that enables precise single-base conversions without cutting both strands of DNA, dramatically improving editing precision. Traditional CRISPR-Cas9 genome editing relies on inducing double-strand breaks in DNA to make targeted changes. While effective, this approach can lead to unintended consequences such as insertions or deletions (referred to as indels) and chromosomal rearrangements. CRISPR base editing has opened doors enabling scientists to correct point mutations linked to genetic diseases without the unintended consequences, leading to companies leveraging base editors to develop cell and gene therapies to help save lives.

What are CRISPR base editors?

A base editor is a gene editing tool that enables single-base changes without creating double-stranded breaks. Each CRISPR base editor generally consists of 3 main components:

  • Modified Cas9 variant such as a nickase (nCas9) or dead Cas9 (dCas9)
  • Deaminase enzyme
  • Base editing guide RNA (gRNA)

A modified Cas9 variant linked to the deaminase enzyme uses the base editing gRNA to guide the base editor to the target site to bind to the DNA without cutting the nucleic acid strand. Once bound to the target site, the deaminase then chemically modifies the target base. Subsequent DNA replication or DNA repair incorporates the new ‘single-letter’ base change. Together, these components enable base editors to efficiently make targeted base conversions to address genetic disease-related mutations like creating termination codons to knock out genes or performing nucleotide changes to correct SNVs, making them powerful tools in a variety of CRISPR applications.

Why Use a Modified Cas9 Variant

Base editing relies on the fusion of a deaminase enzyme with a catalytically impaired variant of a Cas nuclease, typically Cas9 or Cas12. The catalytically impaired Cas protein is engineered to retain its DNA-binding but is rendered incapable of introducing double-strand breaks. Instead, it precisely positions the deaminase to act on a target base within a specific DNA sequence. The Cas protein’s function in this system is purely to facilitate sequence targeting and spatial positioning, ensuring that the deaminase can act with high precision at the intended site within the genome.

Dead Cas9 (dCas9) and Cas9 nickase (nCas9) are widely used in base editing. dCas9 is a catalytically inactive Cas9 variant that binds DNA with high precision, guided by a base editing gRNA molecule, but lacks nuclease activity. This allows it to act solely as a DNA-binding scaffold, positioning the fused deaminase enzyme directly at the target site. Cas9 nickase, on the other hand, retains one active nuclease domain, introducing a single-strand nick instead of a cut through both strands. While this nick can enhance the DNA repair process, increasing the efficiency of the intended base conversion, it can also result in unintended insertions or deletions (indels) at the repair site, which may compromise editing precision. Both variants, however, remain crucial components of base editors for achieving precise and controlled genetic modifications.

What is the Function of the Deaminase?

Once bound to the target site, the linked deaminase chemically modifies the target base. A deaminase is a type of enzyme that removes an amino group (-NH2) from a molecule, a process known as deamination. These enzymes play a critical role in various biological pathways by modifying specific nucleotides in DNA or RNA, effectively altering their chemical structure. For instance, certain deaminases, like cytidine deaminases, can convert a cytosine (C) base into uracil (U) in RNA or into a uracil-like intermediate in DNA. This targeted chemical change creates opportunities to rewrite genetic information without physically cutting the DNA, making deaminases powerful molecular biology tools.

The deaminase enzyme is physically incorporated into the base editor complex by fusing it to the catalytically impaired Cas protein, typically at specific termini such as the N- or C-terminal regions. This strategic placement is crucial to ensure proper spatial alignment between the enzyme and the target DNA, allowing the deaminase to access and modify the desired nucleotide. However, this integration poses structural challenges, as the fusion must maintain the stability, activity, and targeting precision of both the catalytically impaired Cas protein and the deaminase. Improper configuration can hinder the complex's functionality or reduce editing efficiency. On the other hand, when effectively designed, this fusion creates a precise and compact system that combines the targeting power of the Cas protein with the enzymatic activity of the deaminase, enabling highly localized and efficient base modifications.

What Makes a Base Editing gRNA?

A guide RNA (gRNA) for base editing requires specific design criteria to ensure precision and efficiency, distinguishing it from gRNAs used for Cas9-mediated knockouts. With base editing, the gRNA must precisely target the site where the desired nucleotide modification occurs, positioning it within the editing window of the deaminase-Cas fusion complex. Unlike gRNAs for knockouts, where the primary goal is to induce a double-strand break at or near a functional gene region, base editing gRNAs prioritize accurate placement of the catalytically impaired Cas protein to ensure efficient and specific catalytic activity by the deaminase enzyme.

The editing window typically spans a narrow range of bases in the protospacer region, with the exact position depending on the molecular architecture of the base editor. The precise positioning of the target base within this window is critical for efficient editing, as bases outside this range cannot be converted effectively. Additionally, strict PAM requirements, dictated by the Cas nuclease component, further constrain the targetable regions and influence the initial binding and targeting of the base editor complex. Off-target potential and the risk of unintended bystander edits within the editing window add further complexity, requiring careful design to minimize such events. Aligning these considerations demands thorough in silico testing and iterative refinement of base editor gRNAs, ensuring they are both accurately positioned, and are compatible with the structural and functional parameters of the base editing system.

Is base editing and prime editing the same thing?

While both base editing and prime editing are precise CRISPR-based gene editing tools, base editing and prime editing are distinct tools with different capabilities. Prime editing also uses a modified Cas9 variant, like Cas9 nickase. However, it goes beyond base editing by enabling all 12 possible base conversions, along with various size insertions and deletions without introducing double-stranded DNA breaks or relying on a donor template. It holds great promise for therapeutic applications, though it comes with its own technical considerations. Interested in learning more about prime editing? Explore it more on our prime editing guide webpage.

Is base editing and prime editing the same thing?

Types of base editors

There are two primary types of CRISPR base editors: cytosine base editors (CBE) and adenine base editors (ABE). Each base editor is designed to perform different base conversions, expanding the range of correctable mutations without introducing double-strand breaks.

Cytosine Base Editors (CBEs)

CBEs convert a cytosine (C) to a thymine (T), resulting in a C•G to T•A substitution. This base conversion is typically produced by the common cytidine deaminase, APOBEC1, fused to a Cas9 nickase. The base editor gRNA directs the CBE to the target site, unwinding the double-stranded DNA and exposing a single-stranded DNA region where the target cytosine is located. In this exposed region, the APOBEC1 deaminase catalyzes the conversion of cytosine to uracil through deamination. This intermediate uracil is structurally similar to thymine and pairs with adenine during DNA replication. In subsequent rounds of DNA synthesis, the uracil is interpreted as thymine by the replication machinery, effectively completing the C•G to T•A base substitution.

The use of uracil as an intermediate is critical to the editing process because direct enzymatic conversion of cytosine to thymine is chemically unfavorable and not feasible with current enzyme systems. By leveraging the natural deamination pathway, the CBE efficiently induces a transition mutation without creating double-stranded breaks. To enhance editing efficiency and prevent cellular repair mechanisms from reversing the change, cytosine base editors also include uracil glycosylase inhibitors (UGI). These inhibit the base excision repair enzyme uracil N-glycosylase (UNG), which would normally recognize and remove uracil from DNA, restoring the original cytosine. The presence of UGI ensures the uracil remains in place long enough to be replicated as thymine in subsequent cell divisions, preventing the cells from repairing the edit back to a cytosine. The original cytosine base editor, called BE3, was published by Komor et al. in 2016 and demonstrated efficient C-to-T conversions in mammalian cells.

Adenine Base Editors (ABEs)

Adenine base editors convert adenine (A) to guanine (G), effectively performing an A•T to G•C substitution. Different from the original CBE reported in 2016, there was no known natural enzyme that deaminates adenine in DNA at the time, they only acted on RNA. To overcome this, researchers needed to engineer the E. coli tRNA adenosine deaminase, TadA. To function on DNA, this engineered TadA variant forms a heterodimer with wild-type TadA and is fused to the Cas9 nickase in the ABE complex. Once the ABE complex binds to DNA, the DNA is unwound exposing the targeted adenine. The ABE deaminase then converts adenine into inosine (I), which the cell interprets as guanine during replication, producing an A•T to G•C substitution. The first ABE, known as ABE7.10, was reported by Gaudelli et al. in 2017, which showed high-efficiency editing at multiple genomic sites.

Each base editor is guided to the target site by a base editor gRNA, which results in the base conversion by the deaminase enzyme. By choosing the appropriate base editor and base editing guide RNA, cytosine and adenine base editors could address a large number of disease-causing point mutations. This has led researchers to incorporate base editors in applications such as cell and gene therapies.

Engineered Variants to Meet Challenges

Base editing technology has revolutionized gene editing, offering single-base resolution for targeted therapy and research. However, its adoption is not without challenges. Researchers face complexities such as off-target effects that can compromise precision, varying editing efficiencies across different contexts, and the need for optimized delivery methods to ensure accurate application. Addressing these hurdles is critical to unlocking the full potential of base editing and advancing its effectiveness across diverse applications.

What Are Bystander Edits?

Bystander edits represent a critical challenge in the field of precision gene editing, particularly in applications requiring high accuracy, such as therapeutic use. Bystander edits are unintended nucleotide changes that occur within the editing window of a base editor, often due to the enzyme’s activity on nearby bases rather than solely at the intended target site. The consequences of bystander edits can vary significantly depending on the genomic context and the coding frame involved. For instance, they may result in silent mutations that do not alter the encoded protein, missense mutations that substitute one amino acid for another, or more severe deleterious mutations that could disrupt protein function. For example, a bystander edit in a coding region could destabilize a protein or, in regulatory regions, alter gene expression unpredictably - compromising both research outcomes and therapeutic safety.

Bystander edits are particularly problematic in therapeutic gene editing - correcting a pathogenic mutation in a critical gene must avoid introducing new mutations that could trigger harmful side effects or unintended consequences in the patient. Several factors contribute to the likelihood of bystander edits, including the sequence composition around the target site, the specificity of the base editing enzyme, and the design of the base editing guide RNA. Additionally, different base editor variants have unique activity profiles, with some displaying broader editing windows that elevate the risk of such unintended changes.

Advances in base editing are helping address these challenges. Enhanced variants such as BE4max and ABE8e exhibit improved specificity and narrower editing windows, significantly reducing the risk of bystander edits. Additionally, computational tools and refined enzyme engineering approaches are enabling more precise targeting and minimizing off-target activity. These innovations are essential for ensuring the safety and reliability of base editing technologies in both research and therapeutic contexts.

What if My Target is Not Near a PAM Site?

The protospacer adjacent motif (PAM), required by the Cas protein component of base editors, plays a critical role in determining where cytosine and adenine base editors can operate. However, this requirement imposes significant constraints on their activity, as each Cas protein recognizes specific PAM sequences. Commonly used base editors with SpCas9 components, for example, are restricted to targets adjacent to 5’-NGG-3’ PAM sites. This limitation creates a narrow activity window, often leaving certain nucleotide positions inaccessible for editing. Consequently, some disease-causing mutations situated away from these PAM sites remain untargetable, hindering potential therapeutic applications.

To address this issue, enhanced Cas variants have been integrated into base editor platforms to expand their utility. SpCas9-NG, for instance, has been engineered to recognize NG PAMs, while xCas9 targets a wider range of sequences, including NGN. These advancements have been incorporated into both cytosine and adenine base editors, allowing precise editing in regions previously blocked by PAM constraints. Additionally, Cas12a (Cpf1), which recognizes T-rich PAMs (TTTV), has been adapted to develop base editors capable of targeting distinct genomic sites unreachable by SpCas9-based systems.

These next-generation base editors have unlocked new possibilities for research and therapeutic applications. With tools like SpCas9-NG and xCas9-base editors, scientists can now address pathogenic mutations in areas previously deemed uneditable, significantly broadening the scope of gene editing. This progress is critical for developing more effective treatments for genetic disorders and reinforces the promise of base editing as a versatile and precise tool in genome editing technologies.

Are Base Editors too Big for Therapeutic Delivery Methods?

Base editors are significantly larger than traditional CRISPR systems due to the fusion of a deaminase enzyme with a catalytically impaired Cas protein. This increased size presents a major challenge for delivery, particularly with adeno-associated viruses (AAVs), which have a limited packaging capacity of approximately 4.7 kilobases. The size of base editors often exceeds this limitation, making AAV-mediated delivery less feasible for therapeutic applications.

To overcome this obstacle, researchers have developed innovative strategies to enable the efficient delivery of base editors. One such approach involves the use of dual-vector systems, which split the base editor components between two separate AAVs that reassemble in target cells. Additionally, non-viral lipid nanoparticles (LNPs) have gained traction as an alternative delivery method, offering the ability to encapsulate larger payloads and reduce the risk of immune responses. Another promising solution is the engineering of compact base editors, such as leveraging Cas variants like Cas9-ms, which are specifically designed to reduce size while maintaining editing efficiency.

These advancements are crucial for tailoring delivery strategies to address therapeutic needs. Compact base editors and innovative delivery approaches like LNPs not only improve delivery efficiency but also minimize immune reactions and enhance targeting precision at specific genomic sites. These solutions are actively expanding the potential of base editing for clinical applications, enabling researchers to tackle genetic disorders with greater flexibility and efficacy.

What Other Challenges Can Affect Base Editing?

Base editing efficiency is impacted by factors such as chromatin accessibility and cell-type-specific DNA repair pathways. Tightly packed heterochromatin regions can limit the ability of base editors to access certain genomic sites, significantly reducing editing efficiency. This poses a particular challenge when targeting genes in these inaccessible areas for research or therapeutic purposes.

Cell-specific DNA repair mechanisms, such as base excision repair (BER) and mismatch repair (MMR), further complicate base editing. These pathways can interact unpredictably with editing intermediates, leading to variability in outcomes or, in some cases, unintended insertions or deletions (indels). These effects highlight the importance of refining base editor tools to achieve more consistent and precise results, especially for therapeutic applications.

To address these challenges, researchers are developing strategies to enhance accessibility and manage DNA repair pathway interactions. Chromatin-modifying compounds, for instance, are being explored to open tightly packed heterochromatin and provide base editors better access to their target sites. Additionally, approaches to modulate DNA repair pathways, such as using small molecules to inhibit BER or MMR activity, are being tested to create more predictable and controlled editing outcomes. These advancements are critical for expanding the effectiveness and precision of base editing tools in both research and clinical settings.

Designing Base Editor gRNA

Successful CRISPR base editing relies heavily on the careful design of the base editor gRNA. When designing gRNA for base editing, you must keep in mind the desired target cytosine or adenine to edit within the editing window. Not properly designing your base editing gRNA could result in missed edits, unwanted bystander edits, and unsuccessful base editing results.

Also, as part of the base editor gRNA design, you must consider the protospacer adjacent motif (PAM) for the Cas nuclease component of the base editor. Most CRISPR base editors have a dead Cas9 (dCas9), nickase Cas9 (nCas9), or other Cas9 variant as part of the base editing component. The traditional dCas9 and nCas9 are SpCas9 variants from Streptococcus pyogenes, which means the PAM is 5’-NGG-3’. If your base editor uses other Cas nuclease variants like Cas12, you must consider that Cas nucleases' PAM in your base editor gRNA design.

Various computational tools can be utilized to assist with designing base editor gRNA. These tools can offer predictive scoring models that evaluate the efficacy of the base editor gRNA by looking at potential editing efficiency, editing window alignment, and off-targets to name a few. Below is a list of some base editing gRNA design tools:

Tool Name Purpose Link
BE-Designer Design optimal gRNAs for cytosine and adenine base editors BE-Designer
BE-Hive Predict editing efficiency and off-target edits for base editors BE-Hive
DeepBaseEditor Deep learning-based prediction of base editing outcomes DeepBaseEditor
BE-DICT Predict off-target deamination sites specific to base editors BE-DICT
CRISPOR Evaluate on-target scores and predict potential off-target sites CRISPOR
CCTop Identify off-target sites for gRNAs in base editing experiments CCTop

By leveraging these predictive models, scientists could successfully design CRISPR base editor gRNA for applications like cell and gene therapies. For base editor therapeutics, sourcing your gRNA from RUO-to-GMP matters to ensure continuity in manufacturing while you scale up and work towards clinical trials.

Early development of a base editor therapeutic demands the use of best-in-class Research sgRNA to ensure precise, efficient, and reliable edits from the start.

As your base editing program scales toward clinical trials, using best-in-class GMP gRNA is critical for maximizing editing fidelity, increased safety, and meeting regulatory standards for the clinic.

Base Editing in CRISPR-Based Therapeutics

With a vast number of known genetic diseases caused by single-base substitution mutations, CRISPR base editors can be ideal candidates for gene editing therapies to correct those mutations. Therapeutic companies are addressing those single-base mutation diseases by employing adenine base editors, cytosine base editors, or other engineered base editors with high-quality base editing gRNA. Below are a few of those companies utilizing base editors in their clinical pipelines.

Key Clinical Trials Utilizing Base Editing

Base Therapeutics

Base Therapeutics is developing NK510, a therapy for advanced solid tumors. The therapeutic rationale behind NK510 is to engineer base edited Natural Killer (NK) cells that enhance tumor targeting while minimizing off-target effects, a critical need in oncology. NK510 uses the AccuBase cytosine base editor and has received IND approval from both the U.S. FDA and China’s NMPA.

Verve Therapeutics

Verve Therapeutics is using an adenine base editor (ABE) to inactivate PCSK9, in a therapeutic molecule called VERVE-102. To deliver VERVE-102, they developed a proprietary GalNAc-lipid nanoparticle (LNP), which is different than the one used in the previous VERVE-101 treatment. VERVE-102 is part of the Heart-2 clinical trials to treat heterozygous familial hypercholesterolemia (HeFH) and premature coronary artery disease (CAD).

Beam Therapeutics

Beam Therapeutics was founded in 2017 by David Liu and Keith Joung which focuses on using adenine base editors and cytosine base editors to treat genetic diseases. Two notable therapies in Beam’s pipeline are BEAM-201 and BEAM-302. BEAM-201 is an allogeneic, quadruple-edited anit-CD7 CAR-T cell therapy designed to treat Relapsed/Refractory T-Cell Acute Lymphoblastic Leukemia (T-ALL) or T-Cell Lymphoblastic Lymphoma (T-LL) by knocking out four key genes, CD7, TRAC, PDCD1, and CD52. BEAM-302 was developed to treat Alpha-1 antitrypsin deficiency (AATD) using base editors delivered using LNPs to the liver.

Empowering Your Base Editor Therapeutic from Discovery to Clinical Trials

As base editors emerge as powerful CRISPR tools for advancing cell and gene therapies, precision, reliability, and regulatory expertise are critical for reaching clinical trials with confidence. For years, we have partnered with developers to provide comprehensive CRISPR solutions and help them overcome the challenges of therapeutic development. With cutting-edge nucleases, best-in-class gRNAs ranging from research use only (RUO) to GMP, and extensive regulatory expertise, we are your trusted partner in scaling base editing therapies. Together, we can unlock the full potential of CRISPR base editing and shape the future of genetic medicine.

Let’s talk about your base editor pipeline and goals—we’re ready to help you move forward.

Let us be your Guide

We're here to help you find the best CRISPR solution for your project.

Schedule a call today