
Kristofer Mussar is Chief Executive Officer (CEO) of VectorBuilder Inc. and Managing Director of VectorBuilder GmbH, where he leads global operations and strategic growth initiatives.
CREDIT: Kristofer Mussar, VectorBuilder Inc.
CRISPR-based gene editing is driving rapid progress in drug discovery, therapeutic development, and fundamental research. As new CRISPR-based therapies move toward clinical application, efforts to optimize and expand the platform have become a key priority.
Much of the conversation still centers on the editing machinery itself, including Cas nucleases and guide RNAs. However, the success of any CRISPR experiment depends just as much on how these components are delivered. Achieving efficient, precise, and reproducible gene editing requires delivery strategies that are carefully tailored to each experimental context, especially when working with difficult-to-transfect cell types commonly used in drug discovery workflows.
Why CRISPR delivery optimization matters
Successful CRISPR gene editing depends on the efficient delivery of key molecular components such as Cas proteins, guide RNAs (gRNAs), and DNA templates to the target cells. A well-optimized delivery strategy improves transfection efficiency, reduces off-target effects, minimizes cellular toxicity, and increases reproducibility. Despite its importance, delivery remains a major challenge, particularly when working with cell types that are difficult to transfect. These include primary immune cells, stem cells, and neurons, which are often less responsive to conventional delivery methods.
The CRISPR-Cas9 system is a widely used genome editing tool derived from a bacterial adaptive immune mechanism. It enables researchers to make targeted modifications to DNA with high precision. The system consists of two key components: the Cas9 endonuclease and a guide RNA (gRNA). The gRNA directs Cas9 to a specific location in the genome by binding to a complementary DNA sequence. Cas9 then introduces a double-stranded break (DSB) at that site. Once the break is made, the cell engages one of two primary DNA repair pathways:
- Non-homologous end joining (NHEJ): This is the predominant repair mechanism in most cell types. It often introduces small insertions or deletions (indels), which can disrupt gene function.
- Homology-directed repair (HDR): This more precise pathway uses a donor DNA template to introduce defined changes, such as point mutations or the insertion of larger sequences, including fluorescent tags or selection markers.
Optimizing the delivery of CRISPR components plays a crucial role in determining which DNA repair pathway is activated. This is particularly important when precise edits are needed, as successful HDR depends on efficient delivery of both the CRISPR machinery and the donor template.
Comparing viral and non-viral CRISPR delivery methods
CRISPR delivery strategies generally fall into two categories: viral and non-viral. Each offers distinct advantages and limitations relevant to drug discovery.
Viral vectors such as lentivirus, adeno-associated virus (AAV), and adenovirus are valued for their high transduction efficiency and sustained gene expression. Lentiviruses infect both dividing and non-dividing cells, making them useful in primary and stem cells. AAVs are known for their favorable safety profile and low immunogenicity, which makes them well-suited for in vivo studies. However, limitations such as restricted packaging capacity, particularly for larger Cas variants, and the risk of immune activation remain significant challenges.
Non-viral methods like electroporation, lipofection, and nanoparticles provide more flexibility and fewer biosafety concerns. They also support larger payloads. Electroporation is particularly effective for hard-to-transfect cells, including primary T cells and hematopoietic stem cells, though it can cause stress and reduced viability if not optimized.
Lipid nanoparticles (LNPs), widely used in mRNA vaccine platforms, offer scalable production, tunable formulation, and low immunogenicity. Still, achieving consistent editing efficiency across cell types remains difficult and often requires extensive optimization.
Tailoring delivery strategies to cell types
CRISPR delivery is highly cell-type dependent, requiring approaches tailored to the unique characteristics of each system. In stem cell research, including induced pluripotent stem cells (iPSCs) and hematopoietic stem cells (HSCs), delivery methods must preserve viability, differentiation potential, and genomic integrity. Lentiviral vectors and electroporation are commonly used for their reliability and efficiency. With electroporation, optimizing pulse parameters can significantly improve survival and editing outcomes.
Immune cells, such as T cells used in CAR-T therapies, are sensitive to manipulation and often show limited proliferation ex vivo. Non-viral methods like electroporation or transient RNA delivery frequently outperform viral systems in these cells, reducing cytotoxicity and enabling quicker recovery, which is important for clinical translation and manufacturing.
Cas9 selection and vector design considerations
Selecting the appropriate Cas9 variant is a critical step in optimizing CRISPR delivery. Streptococcus pyogenes Cas9 (SpCas9) remains the most commonly used variant due to its high efficiency and well-established protocols. However, alternatives such as Staphylococcus aureus Cas9 (SaCas9) are often better suited for use with AAV vectors, as their smaller size makes them easier to package within the vector's limited capacity. Other engineered tools — such as Cas9 nickases (e.g., D10A), base editors, and prime editors — provide increased specificity or precision but may require dual guide RNAs or more sophisticated delivery strategies.
Vector design also plays a central role in the success of CRISPR experiments. One key decision is whether to use an all-in-one vector that co-expresses both Cas9 and the gRNA, or to separate these components into individual vectors. All-in-one vectors streamline the delivery process, reducing variability and improving co-expression. However, their utility can be limited by vector size constraints. In contrast, separate vectors offer greater flexibility for larger or modular designs and are often used when stable or inducible Cas9 expression is needed.
Additional vector elements can significantly enhance functionality. Fluorescent markers or antibiotic resistance genes assist with the selection and tracking of edited cells. Inducible promoters, cell-type-specific regulatory sequences, and optimized untranslated regions (UTRs) can improve expression control and increase precision, especially in complex or sensitive systems.
Multiplex editing and temporal control
As CRISPR applications expand, drug discovery increasingly relies on advanced capabilities such as multiplex editing and precise temporal control of gene editing. These emerging use cases require delivery systems that can accommodate more complex payloads while maintaining efficiency and specificity.
Multiplex editing involves the simultaneous targeting of multiple genomic loci within the same cell. This approach is essential for interrogating biological pathways, validating drug targets, and modeling polygenic diseases. However, delivering multiple gRNAs and Cas9 constructs in parallel significantly increases payload size and system complexity. Lentiviral vectors and electroporation-based methods are particularly well suited for multiplex applications, as they support the delivery of larger constructs and maintain high editing efficiency across different cell types.
Temporal control allows researchers to regulate the timing of CRISPR activity, enabling edits to occur at specific developmental stages or in response to external cues. This level of control is especially valuable for studying dynamic processes or minimizing cellular stress. Inducible systems and conditional promoters are commonly used to achieve this, with lentiviral vectors providing stable genomic integration and consistent, regulated expression. In contrast, non-viral delivery methods such as electroporation or lipid nanoparticles offer naturally transient expression, which can be advantageous in workflows where temporary activity is preferred.
HDR optimization for precise edits
When HDR is the desired outcome, effective delivery of the donor DNA template is just as critical as delivery of the CRISPR machinery itself. Single-stranded oligodeoxynucleotides (ssODNs) are typically used for introducing small edits or sequence tags under 60 base pairs, while double-stranded DNA (dsDNA) templates enable the insertion of larger elements such as fluorescent reporters or selection markers, often ranging from one to five kilobases.
The choice of delivery method significantly influences HDR efficiency and precision. Electroporation is widely considered the preferred approach for delivering HDR templates, particularly in stem cells and other cell types that are difficult to transfect using conventional methods. Optimizing parameters such as voltage, pulse duration, and DNA concentration can further improve outcomes and minimize cellular stress.
Some platforms, including those offered by VectorBuilder, provide customized donor template design services that align with specific gRNA cut sites. These tailored designs help maximize on-target repair efficiency while reducing the risk of random integration, which is especially important in sensitive cell systems or therapeutic contexts.
New frontiers in CRISPR optimization
As CRISPR delivery systems mature, emerging innovations are pushing the field into new terrain. VectorBuilder is actively developing a range of next-generation tools to further boost precision and safety. These include optimized Cas9 mRNA and sgRNA delivery paired with circular single-stranded DNA (cssDNA) donors, an increasingly preferred format for high-efficiency, non-viral knock-in via HDR. Early-stage research has shown cssDNA can enable knock-in efficiencies of up to 70 percent in iPSCs and improved performance in primary immune cells.
In parallel, the team is pioneering Transcription Activator-Like Effector Nuclease (TALEN) mRNA-based genome editing, which offers complementary specificity benefits to CRISPR and has shown promising results in targeting integrated and episomal hepatitis B DNA.
To support transient, non-integrating editing workflows, VectorBuilder is also advancing lipid nanoparticle (LNP) formulations for the efficient delivery of gene-editing mRNA into hard-to-transfect cells. Their internal proof-of-concept using Enhanced Green Fluorescent Protein (EGFP) mRNA in primary T cells validates the platform’s potential. LNPs are especially attractive for clinical applications given their scalable manufacturing, payload flexibility, and reduced immunogenicity. This is a critical advantage over AAVs, which carry known risks of hepatotoxicity and immune response activation.
Together, these efforts signal a future where CRISPR editing is faster, safer, and more precisely tailored to the demands of modern gene and cell therapy development.
Delivery as a determinant of success
Optimum CRISPR delivery is a core driver of success in gene editing for drug discovery. Achieving consistent, high-efficiency edits across varied cell types demands an integrated approach. This approach must consider delivery modality, cell context, and vector design in equal measure.
As delivery strategies become more refined and adaptable, researchers will be better equipped to harness the full potential of CRISPR. These advances will not only improve experimental reproducibility but also unlock faster drug development, deeper insights into disease biology, and broader therapeutic innovation.
This article was contributed by Kristofer Mussar, CEO of VectorBuilder.









