Tel:
Email:

Enzymatic mRNA Capping for Vaccine and Gene Therapy Development

The rapid rise of messenger RNA (mRNA) technology has completely revolutionized the field of biomedicine. Its core advantage lies in its ability to efficiently guide protein synthesis within cells, offering unprecedented opportunities for the development of new vaccines and precision therapies. However, unmodified "naked" mRNA molecules are extremely unstable in vivo, easily recognized and cleared by the immune system, and have low translation efficiency. The 5'-end cap structure of mRNA molecules, one of the most critical post-transcriptional modifications, plays an indispensable role in overcoming these challenges. It not only effectively shields the mRNA ends from degradation by nucleases, significantly extending the molecule's half-life in vivo, but more importantly, it initiates efficient protein translation by specifically binding to the eukaryotic translation initiation factor 4E (eIF4E). Additionally, specific capping forms (such as Cap 1) can effectively reduce the risk of mRNA molecules being mistakenly identified by intracellular pattern recognition receptors (such as RIG-I) as exogenous danger signals (such as viral RNA), thereby significantly reducing unnecessary immunogenicity. Therefore, achieving efficient, precise, and industrially scalable mRNA capping has become the core technological foundation for ensuring the safety and efficacy of mRNA drugs. Enzyme-mediated capping technology, which precisely synthesizes cap structures in vitro by mimicking the natural cellular pathway, is emerging as the foundational process for the development of next-generation mRNA therapies due to its exceptional performance.

Why Enzymatic Capping is Essential?

In the early stages of mRNA technology development, chemical capping (such as co-transcriptional capping analogues) was the mainstream method. However, this method has insurmountable limitations: low capping efficiency, prone to forming directionally incorrect reverse cap structures, difficulty in efficiently generating advanced Cap 1 structures, and poor cost-effectiveness in large-scale production. Enzyme-mediated mRNA capping technology, which mimics the natural biosynthetic pathways within cells, utilizes recombinant expression of key enzymes (such as the vaccinia virus capping enzyme complex and 2'-O-methyltransferase) to catalyze the capping process stepwise in vitro. This technology has become the core process for the production of next-generation mRNA therapeutic products, with its core advantages lying in its high degree of biomimicry, excellent process performance, and good compliance.

Mimics natural cellular process

The most significant advantage of enzyme-catalyzed capping is its ability to mimic natural cellular processes. This process is catalyzed by a series of highly specific enzymes in the cytoplasm: RNA triphosphatase removes the γ-phosphate group from the 5' end of mRNA, forming a diphosphate terminus; subsequently, guanylyltransferase connects guanosine monophosphate (GMP) via a unique 5'-5' triphosphate bond, forming the Cap 0 core structure (m⁷GpppN); Finally, a specific 2'-O-methyltransferase adds a methyl group to the 2'-O position of the ribose on the first nucleotide of the Cap 0 structure, forming the Cap 1 structure (m⁷GpppNm) with immune-silencing properties. The in vitro enzymatic system precisely replicates this biological cascade reaction, ensuring that the resulting cap structure is identical to that of natural mRNA, thereby maximizing the biological function of the mRNA molecule—efficient translation initiation and low immunogenicity. This biocompatibility is critical for therapeutic mRNA to exert its intended effects in the human body.

Higher accuracy & yield

Higher precision and yield are the core process advantages offered by enzymatic capping technology. Compared to chemical capping, enzymatic reactions exhibit extremely high substrate specificity and catalytic efficiency. Modern recombinant engineering techniques enable the efficient expression and purification of key capping enzymes (such as capping enzymes and 2'-O-methyltransferases) in E. coli or insect cell systems, yielding high-activity, high-purity enzyme preparations. An optimized enzymatic reaction system can achieve near-quantitative capping efficiency (typically >99%) and precisely generate the target Cap structure (Cap 0 or Cap 1), effectively avoiding the common byproducts of chemical methods (such as uncapped mRNA, reverse caps, Cap 0, etc.). This not only significantly improves the uniformity and quality of the final mRNA product but also means that more fully functional mRNA molecules can be obtained per unit of raw material input, greatly enhancing the overall yield and economic efficiency of the production process.

GMP-ready workflows

A workflow compliant with GMP standards is the key to ensuring the successful industrialization of enzyme-catalyzed capping technology. This technology naturally aligns with the GMP (Good Manufacturing Practice) standards required for modern biopharmaceutical production and can be easily integrated into large-scale, compliant production workflows. As biological reagents, the production process of key enzymes (fermentation, purification, formulation, quality control) can establish strict GMP standard operating procedures. Enzyme-catalyzed reactions typically occur in mild aqueous buffer systems, with controllable conditions (temperature, pH, time, substrate ratio, etc.), enabling online monitoring and parameter adjustment to ensure high batch-to-batch consistency. After the reaction is complete, the enzyme protein can be efficiently removed using mature chromatography techniques (such as affinity chromatography, ion exchange chromatography) or tangential flow filtration, meeting the stringent requirements for impurity residues in the final product. The robustness, scalability, and controllability of this process lay a solid production foundation for meeting the massive global demand for mRNA vaccines and drugs in the future.

Applications of Enzymatic mRNA Capping

With its comprehensive advantages in high efficiency, high fidelity, low immunogenicity, and compliance with GMP production standards, enzyme-catalyzed capping technology has become a powerful engine driving the development of multiple key therapeutic areas, particularly demonstrating significant application value and broad prospects in the fields of infectious disease prevention, cancer treatment, and gene therapy repair.

Infectious disease vaccines

Infectious disease vaccines represent the most successful and widely applied field of enzyme-catalyzed capping technology. This technology serves as the cornerstone for developing efficient and safe mRNA vaccines. The tremendous success of COVID-19 mRNA vaccines (such as Pfizer-BioNTech's Comirnaty and Moderna's Spikevax) has fully validated the critical role of the Cap 1 structure in balancing strong immunogenicity (inducing high levels of neutralizing antibodies and T cell responses) with acceptable safety (reducing non-specific inflammatory responses). Enzyme-mediated capping technology enables the large-scale production of mRNA antigen-encoding sequences with a high proportion of Cap 1 structures and high uniformity, ensuring efficient expression of the target antigen (such as the SARS-CoV-2 spike protein) in the human body while minimizing excessive innate immune activation caused by uncapped or Cap 0 mRNA. This technology is being rapidly applied to address other major infectious disease threats, including influenza (pursuing broad-spectrum or universal vaccines), respiratory syncytial virus (RSV), human immunodeficiency virus (HIV), Ebola virus, Zika virus, and future emerging infectious disease pathogens. Its rapid adaptability enables swift initiation of vaccine development upon obtaining pathogen gene sequences, providing a powerful tool for global public health security.

Oncology immunotherapies

Tumor immunotherapy is a key area where enzyme-catalyzed capping technology demonstrates significant potential. The application of mRNA technology in this field primarily includes personalized neoantigen vaccines and therapeutic monoclonal antibody (mAb) encoding mRNA. Personalized neoantigen vaccines are developed by sequencing to identify patient-specific tumor mutations and designing mRNA vaccines that encode these neoantigens. The Cap 1 structure generated by enzyme-catalyzed capping is critical for such vaccines: on one hand, it ensures efficient expression of neoantigens within antigen-presenting cells (such as dendritic cells), thereby effectively activating the patient's own tumor-specific T cell immune response; on the other hand, its low non-specific immunogenicity helps focus the immune response on the target neoantigens, reducing off-target effects, and may allow for higher doses or more frequent administration to enhance efficacy. Additionally, utilizing mRNA for transient expression of therapeutic antibodies (such as immune checkpoint inhibitors targeting PD-1/PD-L1 or CTLA-4, or bispecific antibodies targeting tumor-associated antigens) is an emerging strategy. Enzyme-mediated capping technology ensures that mRNA encoding therapeutic antibodies efficiently and rapidly produces fully functional antibody proteins at therapeutic concentrations in the liver or targeted delivery sites, overcoming issues such as high production costs and potential toxicity accumulation from long half-lives associated with recombinant protein antibodies. This provides more flexible and controllable options for cancer treatment.

Cell & gene therapy development

The development of cell and gene therapies is an increasingly important application area for enzyme-catalyzed capping technology. Its applications primarily manifest in two aspects: first, it is used for in vitro transcription (IVT) to prepare mRNA encoding gene editing tools (such as CRISPR-Cas9 nucleases, base editors, and guide RNAs). Delivering mRNA encoding Cas9 protein and sgRNA into in vitro cultured cells (such as T cells, hematopoietic stem cells, and induced pluripotent stem cells (iPSCs)) via methods like electroporation enables efficient gene editing (such as knockout, knock-in, and base substitution). The enzyme-catalyzed capping of the Cap 1 structure is critical for such edited mRNA, significantly enhancing the intracellular expression efficiency and precision of editing elements (reducing off-target editing or cellular damage caused by immune activation), while reducing cellular toxicity caused by uncapped RNA. This significantly improves the safety and efficiency of gene editing, making it a key upstream raw material for producing engineered cells for CAR-T cell therapy or genetic disease gene correction. Second, it is used for in vivo gene therapy or protein replacement therapy. Through lipid nanoparticles (LNP) or other delivery vehicles, mRNA encoding functional proteins (such as enzymes or secreted proteins to treat genetic diseases) is directly delivered to the target organs (such as the liver) of patients. Enzyme-mediated capping technology ensures that such therapeutic mRNA can achieve high-level and sustained protein production within target cells while minimizing the risk of systemic inflammatory responses. This provides an important alternative or complementary approach to traditional gene therapy (such as viral vectors), particularly for scenarios requiring transient expression or repeatable administration.

Our Enzymatic Capping Capabilities

To meet the urgent demand for high-quality capping technology in the field of mRNA therapy, we have established a comprehensive and advanced enzyme-catalyzed capping platform focused on providing efficient, precise, and compliant capping solutions covering the entire chain from core enzyme development to final product quality control. The core of this platform lies in the precise synthesis of key cap structures, in-depth optimization of core enzymes, and strict quality control throughout the entire process.

m⁷G and Cap 1 structures

The efficient and precise synthesis of m⁷G and Cap 1 structures is the core output of our platform. We utilize a thoroughly researched and engineered vaccinia virus capping enzyme system. This system consists of two subunits: the D1 subunit (possessing dual activity as an RNA triphosphatase and guanylyltransferase) and the D12 subunit (acting as a regulatory subunit to enhance D1 activity). This complex efficiently catalyzes the conversion from 5'-triphosphate RNA (ppp-RNA) to the Cap 0 structure (m⁷GpppN) in a single step. More importantly, our platform integrates highly active recombinant 2'-O-methyltransferases (such as the VP39 protein from the vaccinia virus or its optimized variants), which can specifically recognize the Cap 0 structure and efficiently convert it into the Cap 1 structure (m⁷GpppNm) with immune silencing advantages. By precisely controlling key parameters such as the ratio of the two enzymes in the reaction system, reaction time, and substrate concentration (e.g., S-adenosylmethionine, SAM), we can achieve high-yield production of the Cap 1 structure, meeting the stringent requirements for low immunogenicity in therapeutic-grade mRNA.

Methyltransferase optimization

Methyl transferase optimization is a decisive step in ensuring the generation of high-quality Cap 1 structures. We have conducted in-depth optimization of the core 2'-O-methyltransferase through a multi-level enzyme engineering strategy. Using directed evolution technology, we performed high-throughput screening targeting key enzymatic properties (such as affinity for Cap 0 substrate (Km), catalytic efficiency (kcat), utilization of SAM, thermal stability, and soluble expression levels), resulting in mutants with significantly improved performance. Concurrently, we analyzed the enzyme-substrate complex using structural biology to guide rational design, aiming to optimize the binding pockets for the enzyme, Cap 0 structure, and methyl donor SAM, thereby reducing non-specific binding and enhancing the specificity and rate of the methylation reaction. These optimization efforts significantly enhance the specific activity of the methyltransferase and its stability under large-scale production conditions, reduce enzyme usage and production costs, while ensuring high efficiency and specificity in the generation of the Cap 1 structure.

Quality control validation

Quality control validation is the lifeline of the entire enzymatic capping process, ensuring the safety and efficacy of the final product. We have established a multidimensional quality control (QC) system that complies with GMP principles. Core technologies include: 1. Liquid chromatography-tandem mass spectrometry (LC-MS/MS): As the gold standard method, it precisely determines the chemical composition of the mRNA 5' end cap structure (clearly distinguishing between Cap 0, Cap 1, Cap 2, etc.), methylation status (m⁷G and Nm), and detects any incomplete or abnormal capping products, providing precise molecular-level information. 2. Capillary Electrophoresis (CE) or High-Performance Liquid Chromatography (HPLC): Used to comprehensively assess the overall integrity and purity of mRNA, confirming that the capping reaction process has not caused significant damage to the mRNA chain (e.g., degradation), and effectively removing residual nucleotides, enzymes, proteins, and small-molecule substrates (e.g., SAM and its metabolite SAH) from the reaction system. 3. Functional validation: Quantitatively assess the translation efficiency of capped mRNA using an in vitro cell-free translation system (such as rabbit reticulocyte lysate or wheat germ extract); evaluate its immunostimulatory properties using specific immune cell lines (such as dendritic cells, macrophages) or carefully designed reporter gene systems to ensure that the Cap 1 structure effectively reduces unnecessary immunogenicity. This rigorous, multi-tiered QC system is integrated throughout the entire process development and production workflow, providing reliable assurance for the critical quality attributes (CQAs) of each batch of capped mRNA products.

Enzyme-mediated mRNA capping technology, which precisely mimics the natural capping pathway of eukaryotic cells, utilizes a highly optimized enzyme system to efficiently synthesize the critical Cap 0 and Cap 1 structures in vitro, has become an indispensable core process in modern mRNA therapeutic platforms. Its core value lies in overcoming the inherent limitations of early chemical capping methods, achieving near-quantitative capping efficiency, exceptional structural fidelity (particularly the generation of a high proportion of Cap 1), and natural low immunogenicity. These characteristics directly translate into longer half-lives, more efficient protein translation capabilities, and significantly improved safety profiles for therapeutic mRNA molecules in the human body.

With the continuous advancement of recombinant protein engineering technology, particularly the directed evolution and rational design of capping enzymes and methyltransferases, the catalytic efficiency, specificity, stability, and cost-effectiveness of enzyme-catalyzed capping systems have been continuously optimized. Concurrently, complementary processes compliant with stringent GMP standards and a multi-dimensional quality control system incorporating cutting-edge technologies such as LC-MS/MS ensure batch-to-batch consistency and high regulatory compliance of mRNA products in large-scale production. This technical maturity and standardization have paved the way for mRNA drugs to transition from the laboratory to industrial-scale production.

At the application level, the enabling effects of enzyme-catalyzed capping technology have become evident in multiple cutting-edge fields of biomedicine. It forms the cornerstone of the success of the next generation of infectious disease mRNA vaccines (such as the COVID-19 vaccine), playing a key role in balancing their strong immunogenicity with good tolerability. In cancer immunotherapy, it drives the precise immune activation of personalized neoantigen vaccines and the efficient in vivo production of therapeutic antibody-encoding mRNA. In the field of cell and gene therapy, it serves as the core technological safeguard for the safe and efficient preparation of gene editing tool mRNA and the realization of in vivo protein replacement therapy.

Looking ahead, as mRNA technology expands into broader disease areas (such as autoimmune diseases, regenerative medicine, and protein replacement therapy) and new delivery systems are developed, the demand for high-quality, low-cost, scalable enzymatic capping technology will only continue to grow. Continuously optimizing enzyme performance, simplifying process workflows, deepening understanding of the biological functions of cap structures, and exploring the potential of novel cap analogues will be key directions for ongoing innovation in this field. Undoubtedly, enzymatic capping technology, as a foundational process in mRNA drug development, will continue to drive revolutionary advancements in the fields of vaccines and gene therapy, providing more powerful tools to combat major human diseases.

Request a Quote for Enzymatic Capping Services

Advance your vaccines and gene therapies with our enzymatic capping solutions. Mimicking natural biological processes, enzymatic capping delivers maximum accuracy, stability, and translation efficiency—critical for high-stakes applications. Our services include Cap 0 and Cap 1 structures, methyltransferase optimization, and GMP-ready processes, ensuring your capped mRNA meets strict regulatory and clinical demands. We support both infectious disease vaccines and oncology immunotherapies, as well as cutting-edge gene therapy projects.

With scalable workflows, strict QC, and expert support, we ensure your RNA is optimized at every stage, from early discovery to clinical production. Clients worldwide rely on us for reliability and scientific precision.

Contact us today for a customized quote and see how our enzymatic capping services can strengthen your vaccine and gene therapy pipelines.

Complementary mRNA Capping Services

mRNA Capping Knowledge Center

* Only for research. Not suitable for any diagnostic or therapeutic use.
Online Inquiry
Verification code
Inquiry Basket