Appearance
This report is written by MaltSci based on the latest literature and research findings
How are mRNA vaccines developed?
Abstract
The emergence of messenger RNA (mRNA) vaccines has marked a transformative shift in vaccine technology, characterized by their ability to rapidly elicit immune responses against infectious diseases. This review explores the historical development, mechanisms of action, design, synthesis, preclinical and clinical development, regulatory pathways, and future perspectives of mRNA vaccines. Initially developed in the 1990s, mRNA technology gained prominence during the COVID-19 pandemic, demonstrating unprecedented efficacy and safety in emergency use scenarios. The mRNA vaccine development process involves the identification of target pathogens, synthesis of mRNA through in vitro transcription, and encapsulation in lipid nanoparticles for effective delivery into host cells. The dual activation of both humoral and cellular immune responses positions mRNA vaccines as a powerful tool against infectious diseases. Regulatory frameworks have evolved to ensure the safety and efficacy of mRNA vaccines, although manufacturing challenges persist. Looking ahead, mRNA technology holds promise not only for infectious disease prevention but also for therapeutic applications in cancer and other conditions. This review underscores the potential of mRNA vaccines to address ongoing and future public health challenges, emphasizing the need for continued innovation and research in this rapidly advancing field.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Background of mRNA Technology
- 2.1 Historical Development of mRNA Vaccines
- 2.2 Mechanism of Action of mRNA Vaccines
- 3 Design and Synthesis of mRNA Vaccines
- 3.1 mRNA Sequence Design
- 3.2 In Vitro Transcription Methods
- 4 Preclinical and Clinical Development
- 4.1 Preclinical Testing in Animal Models
- 4.2 Phases of Clinical Trials
- 5 Regulatory Approval and Manufacturing Challenges
- 5.1 Regulatory Pathways for mRNA Vaccines
- 5.2 Manufacturing and Quality Control Issues
- 6 Future Perspectives and Applications
- 6.1 Potential for Other Infectious Diseases
- 6.2 Therapeutic Applications Beyond Vaccines
- 7 Conclusion
1 Introduction
The emergence of messenger RNA (mRNA) vaccines marks a revolutionary shift in the landscape of vaccine technology and immunology. Historically, the development of vaccines has relied on traditional platforms, such as live attenuated or inactivated pathogens, which often require extensive time and resources to produce. In contrast, mRNA vaccines harness the body’s own cellular machinery to generate immune responses, representing a paradigm shift that enables rapid development and deployment against infectious diseases. The successful introduction of mRNA vaccines during the COVID-19 pandemic has not only demonstrated their efficacy but also underscored their potential for broader applications in infectious diseases, cancer treatment, and beyond [1][2].
The significance of mRNA vaccines extends beyond their immediate applications. Their rapid development during the COVID-19 pandemic has showcased an unprecedented capability to respond to emerging health crises, providing a robust framework for future vaccine development. This is particularly crucial as the global community faces ongoing threats from novel pathogens and the resurgence of existing infectious diseases [2][3]. The flexibility of mRNA technology allows for the quick adaptation of vaccine designs to match the evolving genetic landscape of pathogens, which is vital in the context of rapidly mutating viruses [4].
Current research highlights the advancements in mRNA technology, including the mechanisms of action, design, synthesis, and delivery of mRNA vaccines. The historical context of mRNA vaccine development dates back to the 1990s, but it was not until the recent pandemic that these technologies were catalyzed into mainstream use [1]. The mechanism by which mRNA vaccines induce an immune response involves the delivery of synthetic mRNA that encodes for specific antigens, prompting the host cells to produce these antigens and elicit both humoral and cellular immune responses [2][3].
This report aims to provide a comprehensive overview of the entire mRNA vaccine development process, structured around the following key sections:
Background of mRNA Technology: This section will explore the historical development of mRNA vaccines and their underlying mechanisms of action, illustrating how these innovations have evolved over time and the scientific principles that govern their function.
Design and Synthesis of mRNA Vaccines: Here, we will delve into the intricate processes involved in designing mRNA sequences and the in vitro transcription methods used to produce them, emphasizing the importance of sequence optimization and stability [5].
Preclinical and Clinical Development: This section will discuss the stages of vaccine development, including preclinical testing in animal models and the phases of clinical trials that mRNA vaccines undergo to assess their safety and efficacy [6].
Regulatory Approval and Manufacturing Challenges: We will examine the regulatory pathways that mRNA vaccines must navigate for approval, alongside the manufacturing challenges that arise in scaling production to meet global demands [7][8].
Future Perspectives and Applications: Finally, this report will explore the potential applications of mRNA technology beyond infectious diseases, including its role in cancer immunotherapy and the development of personalized medicine [9].
Through this structured analysis, we aim to elucidate the multifaceted journey of mRNA vaccines from conceptualization to public health implementation, contributing to a deeper understanding of how these vaccines can be leveraged to combat future pandemics and improve global health outcomes. As we navigate the complexities of vaccine development in a rapidly changing world, the insights gained from mRNA vaccine technology will be instrumental in shaping the future of immunization strategies.
2 Background of mRNA Technology
2.1 Historical Development of mRNA Vaccines
The development of messenger RNA (mRNA) vaccines represents a significant advancement in vaccine technology, driven by both historical research and recent innovations. The origins of mRNA technology can be traced back to the 1970s, but it gained substantial momentum in the early 1990s when the first studies demonstrated the potential of mRNA as a vaccine platform. The initial focus was on mRNA's ability to induce immune responses against infectious diseases, notably influenza, which was investigated in a study conducted in 1993 (Leong et al. 2025) [1].
The urgency brought on by the COVID-19 pandemic catalyzed the rapid development of mRNA vaccines, leading to unprecedented global collaboration and investment in this technology. Notably, the Pfizer-BioNTech and Moderna vaccines showcased the feasibility and effectiveness of mRNA vaccines, achieving emergency use authorization in record time (Zheng & Feng 2025) [4]. The success of these vaccines has not only addressed the immediate public health crisis but has also underscored the transformative potential of mRNA technology in vaccine development.
The mRNA vaccine development process typically involves several key steps. First, the target pathogen's genomic sequence is identified, allowing researchers to design an mRNA sequence that encodes the target antigen. This mRNA is synthesized through in vitro transcription, a process that allows for precise control over the mRNA's structure and sequence (Li et al. 2025) [2]. The synthesized mRNA is then encapsulated in lipid nanoparticles (LNPs), which serve as delivery vehicles to facilitate the entry of mRNA into host cells (Matarazzo & Bettencourt 2023) [3].
The incorporation of modified nucleotides into the mRNA can enhance stability and reduce innate immune responses, improving the overall efficacy of the vaccine (Wei et al. 2025) [10]. Furthermore, advancements in nanotechnology have played a critical role in optimizing the delivery systems for mRNA vaccines, enabling them to elicit robust immune responses while minimizing potential side effects (Gote et al. 2023) [11].
Historically, the application of mRNA technology faced challenges, including stability issues and the need for efficient delivery mechanisms. However, innovations such as self-amplifying RNA and novel LNP formulations have addressed some of these hurdles, allowing for enhanced antigen expression and improved immunogenicity (Knezevic et al. 2021) [6]. The regulatory landscape has also evolved, with organizations like the World Health Organization (WHO) establishing guidelines to ensure the quality, safety, and efficacy of mRNA vaccines, facilitating international collaboration in vaccine development (Knezevic et al. 2021) [6].
In conclusion, the development of mRNA vaccines is a culmination of decades of research, technological advancements, and a responsive approach to emerging health threats. The rapid deployment of mRNA vaccines during the COVID-19 pandemic has set a precedent for future vaccine development, potentially revolutionizing how vaccines are designed and manufactured for various infectious diseases, including influenza, HIV, and even cancers (Maruggi et al. 2019) [12]. The ongoing evolution of mRNA technology holds promise for addressing global health challenges in the years to come.
2.2 Mechanism of Action of mRNA Vaccines
mRNA vaccines represent a groundbreaking advancement in vaccine technology, emerging from decades of research into messenger RNA (mRNA) and its applications in immunology. The development of mRNA vaccines involves several critical stages, including the design of the mRNA sequence, in vitro transcription, formulation, and delivery mechanisms.
The process begins with the selection of the target antigen, which is typically a protein associated with a pathogen, such as a virus. The corresponding mRNA sequence is then synthesized, usually using a DNA template that encodes the antigen. This step involves in vitro transcription, where RNA polymerase is used to produce mRNA from the DNA template. The mRNA produced is often modified to enhance its stability and translation efficiency, such as by adding a 5' cap and a poly(A) tail, which protect the mRNA from degradation and facilitate its translation into protein once inside host cells[1][10].
Once synthesized, the mRNA is encapsulated in lipid nanoparticles (LNPs) to facilitate its delivery into cells. LNPs serve as carriers that protect the mRNA from degradation in the bloodstream and enhance its uptake by cells. The use of LNPs has been pivotal in the success of mRNA vaccines, as they allow for efficient delivery of the mRNA into the cytoplasm of target cells, where it can be translated into the target protein[2][3].
The mechanism of action of mRNA vaccines is relatively straightforward yet highly effective. After administration, the lipid nanoparticles deliver the mRNA into the cytoplasm of the host cells. The host cell's ribosomes then translate the mRNA into the encoded antigen protein. This protein is subsequently processed and presented on the cell surface by major histocompatibility complex (MHC) molecules, which play a crucial role in the activation of T cells, a key component of the adaptive immune response[4][10].
In addition to eliciting a T-cell response, the expressed antigen can also stimulate B cells to produce antibodies, further enhancing the immune response against the pathogen. This dual activation of both arms of the immune system (humoral and cellular) is a significant advantage of mRNA vaccines, as it can lead to a robust and long-lasting immunity[3][9].
Despite their advantages, the development of mRNA vaccines is not without challenges. Issues such as mRNA stability, the need for cold chain logistics for storage and transport, and the potential for adverse events, including rare occurrences like myocarditis, must be addressed to ensure the safe and effective deployment of these vaccines[8][13]. Furthermore, ongoing research aims to optimize mRNA design and delivery systems to improve efficacy and reduce side effects[2][4].
In conclusion, the development of mRNA vaccines involves a complex interplay of molecular biology techniques, advanced delivery systems, and a thorough understanding of immune mechanisms. This innovative approach not only offers a rapid response to emerging infectious diseases but also holds promise for applications in cancer treatment and other therapeutic areas[5][14].
3 Design and Synthesis of mRNA Vaccines
3.1 mRNA Sequence Design
The development of mRNA vaccines involves a series of meticulously planned steps that begin with the design of the mRNA sequence. This process is crucial for ensuring the efficacy and safety of the resulting vaccine. The following outlines the key stages in the design and synthesis of mRNA vaccines, particularly focusing on mRNA sequence design.
The first step in developing an mRNA vaccine is the specification of the antigen sequence that corresponds to the target antigen. This specification is essential as it guides the design and manufacture of the mRNA vaccine. The target antigen is typically a protein associated with the pathogen or disease of interest, and its sequence must be accurately identified to ensure the vaccine elicits an appropriate immune response[9].
Once the target antigen sequence is established, the next phase involves the isolation of the mRNA from the target protein using a nucleic acid RNA-based vaccine approach. This process includes constructing a DNA template that corresponds to the desired mRNA sequence. The DNA template is then subjected to in vitro transcription, which converts the DNA into mRNA strands. This transcription process is crucial as it allows for the production of mRNA that encodes the target antigen[9].
After transcription, several modifications are made to the mRNA to enhance its stability and translation efficiency. These modifications include the addition of a 5' cap and poly(A) tail to the mRNA. The 5' cap protects the mRNA from degradation and assists in the initiation of translation, while the poly(A) tail improves mRNA stability and enhances the efficiency of translation[9].
Following the synthesis and modification of the mRNA, a purification process is conducted to remove any contaminants that may have been introduced during the preparation. This purification is vital to ensure the quality and safety of the mRNA product before it is formulated into a vaccine[9].
The formulation of the mRNA vaccine involves encapsulating the mRNA in delivery systems, such as lipid nanoparticles (LNPs). These nanoparticles are essential for protecting the mRNA from degradation and facilitating its delivery into host cells. The use of LNPs has been shown to significantly enhance the stability and efficacy of mRNA vaccines, allowing for effective triggering of both adaptive and innate immune responses[9].
Overall, the design and synthesis of mRNA vaccines are characterized by a combination of molecular biology techniques and innovative formulation strategies. The careful consideration of each step, from sequence design to delivery, is crucial for the successful development of effective mRNA vaccines against various diseases[3][15].
3.2 In Vitro Transcription Methods
The development of mRNA vaccines involves several critical steps, particularly in the design and synthesis phases, with in vitro transcription (IVT) methods playing a pivotal role. mRNA vaccines utilize single-stranded linear DNA as a template for the in vitro transcription process, which is essential for producing the mRNA that encodes the target antigen. This antigen, once delivered into host cells, can elicit an immune response by enabling the expression of the target protein [10].
A conventional method for preparing template DNA involves plasmid propagation in bacteria, followed by enzymatic linearization. However, this approach can be labor-intensive and costly. Recent advancements have introduced a cell-free, PCR-based approach for generating high-quality linear DNA templates. This PCR-based method has been shown to yield higher amounts of both DNA templates and transcribed mRNA compared to traditional plasmid-derived methods, while maintaining the quality and integrity of the mRNA produced [16].
In vitro transcription reactions are typically catalyzed by RNA polymerase and require optimization to ensure efficiency and cost-effectiveness. The optimization of IVT conditions can significantly enhance the yield and quality of mRNA. For instance, studies have demonstrated that adjustments in parameters such as temperature, concentration of urea, and the inclusion of reaction-enhancing additives can lead to substantial improvements in mRNA production. One study reported a 55% increase in mRNA yield while reducing truncated mRNA by 33% through optimized reaction conditions [17].
Moreover, the integration of machine learning techniques, specifically Bayesian optimization, has emerged as a powerful tool for refining IVT processes. This approach allows for data-driven optimization of reaction conditions, leading to high yields in a shorter time frame. In one instance, an IVT reaction produced 12 g/L of mRNA in just 2 hours, significantly outperforming industry standards [18].
To support the manufacturing process, analytical methods such as high-performance liquid chromatography (HPLC) have been developed to monitor mRNA production and assess the purification of mRNA products. This method enables rapid analysis and quantification of key components during IVT, ensuring that the final mRNA product meets the required quality standards [19].
Overall, the development of mRNA vaccines encompasses a combination of innovative synthesis methods, optimization of transcription processes, and the implementation of advanced analytical techniques. These advancements not only enhance the efficiency and scalability of mRNA vaccine production but also pave the way for rapid responses to emerging infectious diseases and the potential application of mRNA technology in various therapeutic areas [1].
4 Preclinical and Clinical Development
4.1 Preclinical Testing in Animal Models
The development of mRNA vaccines is a multifaceted process that involves several stages, including preclinical testing in animal models, which is critical for evaluating safety and efficacy before progressing to human clinical trials. Preclinical testing typically employs a variety of animal models to assess the immune response, safety, and overall effectiveness of the vaccine candidates.
Animal models play a crucial role in the development of vaccines, particularly for infants and young children, as they help in assessing the immune response's quantity and quality, the onset and duration of immunity, and the safety and toxicity of the vaccine formulation [20]. In the context of mRNA vaccines, preclinical studies have shown that these vaccines can elicit strong immune responses in various animal models, including rodents and larger animals, which more accurately predict human outcomes [21].
Zebrafish embryos have emerged as a valuable model organism for testing mRNA-based therapeutics due to their small size, optical transparency, and high embryo yield, which facilitate high-throughput drug testing [22]. The one-cell stage injection of naked mRNA can be employed to evaluate gene addition effects in vivo, while intravascular injections in older larvae can assess tissue targeting efficiency of packaged mRNA. This approach allows researchers to gather preliminary data on the efficacy of mRNA vaccines before moving to more complex mammalian models.
The use of large animal models is also emphasized in the preclinical development of vaccines. Regulatory authorities require that novel vaccine candidates undergo extensive preclinical assessment in these models to ensure they can safely and effectively elicit the desired immune response [21]. Large animals, such as pigs and non-human primates, are particularly useful because they provide a more relevant physiological context for predicting human responses compared to smaller models.
As mRNA vaccine technology has advanced, the focus has shifted towards optimizing various components, including mRNA design, delivery systems, and the identification of suitable adjuvants to enhance immunogenicity [23]. Innovations such as lipid nanoparticles (LNPs) have revolutionized the delivery of mRNA, allowing for effective encapsulation and cellular uptake [1]. The preclinical phase also includes extensive safety assessments, as any adverse effects must be identified and mitigated before human trials can commence.
In summary, the development of mRNA vaccines is a comprehensive process that begins with preclinical testing in animal models. These models are essential for understanding the immune response, ensuring safety, and optimizing vaccine formulations before advancing to clinical trials in humans. The ongoing refinement of mRNA vaccine technology continues to enhance its potential in combating infectious diseases and other health challenges.
4.2 Phases of Clinical Trials
The development of mRNA vaccines involves a structured process that includes preclinical and clinical phases, characterized by specific stages of clinical trials designed to evaluate safety, immunogenicity, and efficacy.
Initially, in the preclinical phase, researchers focus on the design and synthesis of the mRNA vaccine. This involves selecting appropriate antigens derived from the target pathogen, followed by in vitro transcription of the mRNA. The synthesized mRNA is then encapsulated in lipid nanoparticles (LNPs) to facilitate delivery into host cells. This stage is critical for assessing the basic safety and immunogenicity of the vaccine before human trials begin[2].
The clinical development of mRNA vaccines is categorized into several phases:
Phase 1 Trials: These trials primarily assess the safety and immunogenicity of the vaccine in a small group of healthy volunteers. The main objectives are to evaluate the vaccine's safety profile and determine the appropriate dosage. For example, the mRNA vaccines developed for COVID-19 demonstrated robust immunogenicity and provided good protection against severe disease during these early trials[24].
Phase 2 Trials: Following successful Phase 1 results, Phase 2 trials involve a larger group of participants. These trials continue to evaluate safety while also providing preliminary data on the vaccine's efficacy. They help in refining the dosage and identifying any side effects that may not have been evident in Phase 1[25].
Phase 3 Trials: These trials are conducted on a much larger scale and are designed to confirm the vaccine's efficacy and monitor its side effects in diverse populations. Phase 3 trials provide the definitive evidence needed for regulatory approval. For instance, the mRNA vaccines for COVID-19 underwent extensive Phase 3 trials, which demonstrated an approximate 85%-95% reduction in the risk of symptomatic COVID-19[26].
Regulatory Approval and Phase 4 Trials: After successful Phase 3 trials, the vaccine is submitted for regulatory approval. Upon approval, Phase 4 trials, also known as post-marketing surveillance, are conducted to monitor the vaccine's long-term safety and effectiveness in the general population. This phase is crucial for identifying rare side effects and assessing the vaccine's performance in real-world settings[27].
Throughout these phases, mRNA vaccines benefit from their "plug and play" technology, allowing rapid adaptation to emerging pathogens and variations in existing viruses, which is particularly important given the high mutation rates of RNA viruses[28]. The mRNA platform's flexibility, combined with the rapid development timelines observed during the COVID-19 pandemic, has accelerated the clinical progress of mRNA vaccines against various infectious diseases[4].
In summary, the development of mRNA vaccines is a meticulous process involving multiple phases of clinical trials that ensure the safety and efficacy of the vaccine before it is made available to the public. The success of mRNA vaccines during the COVID-19 pandemic has paved the way for ongoing research and development against a wide array of infectious diseases[29][30][31].
5 Regulatory Approval and Manufacturing Challenges
5.1 Regulatory Pathways for mRNA Vaccines
The development of mRNA vaccines involves several critical phases, including regulatory approval and addressing manufacturing challenges. The rapid advancements in mRNA vaccine technology, particularly highlighted during the COVID-19 pandemic, have prompted significant regulatory considerations to ensure safety, efficacy, and accessibility.
Regulatory pathways for mRNA vaccines are evolving as the technology matures. The World Health Organization (WHO) has emphasized the need for global norms and standards for biological products, including vaccines, to assure their quality, safety, and efficacy. These efforts are essential to facilitate international convergence of manufacturing and regulatory practices, thereby supporting National Regulatory Authorities in WHO member states [6].
In the context of mRNA vaccine development, the U.S. Food and Drug Administration (FDA) has provided guidance that allows for expedited review processes under emergency use authorizations (EUAs), as seen with the Pfizer-BioNTech and Moderna vaccines. These vaccines received EUAs based on robust clinical trial data demonstrating their safety and efficacy against COVID-19 [2]. The rapid development cycle of mRNA vaccines, which can be designed and produced within days, represents a significant advantage over traditional vaccine platforms [5].
However, the manufacturing of mRNA vaccines presents unique challenges. The production process typically involves in vitro transcription of mRNA, followed by encapsulation in lipid nanoparticles (LNPs) for effective delivery into host cells. Despite the relatively straightforward production methods, significant hurdles remain, including mRNA stability, the need for optimized delivery systems, and ensuring the absence of immunostimulatory byproducts that could hinder long-term therapeutic applications [32].
Moreover, the high costs associated with raw materials, along with the complexities of scaling up production, pose additional challenges. The dependency on global supply chains for quality raw materials and other supplies can also impede timely vaccine development and distribution [33]. The introduction of quality by design (QbD) frameworks and standardized quality control protocols is crucial for maintaining high-quality mRNA vaccines and addressing these manufacturing challenges [8].
In summary, the development of mRNA vaccines encompasses a multifaceted approach involving regulatory frameworks that ensure safety and efficacy, alongside addressing manufacturing challenges that can affect the scalability and accessibility of these vaccines. Continued collaboration between regulatory authorities, manufacturers, and researchers will be essential to fully harness the potential of mRNA vaccine technology in combating infectious diseases.
5.2 Manufacturing and Quality Control Issues
The development of mRNA vaccines involves a multifaceted process that includes regulatory approval and manufacturing challenges, particularly concerning quality control. mRNA vaccines have emerged as a revolutionary approach in modern vaccinology, showcasing their potential in addressing both infectious diseases and cancer therapies.
The manufacturing process of mRNA vaccines typically begins with the design of the mRNA sequence, which encodes the target antigen. This is followed by the preparation of a DNA template through PCR amplification. The subsequent in vitro transcription of this DNA template generates the mRNA, which is then capped and polyadenylated to enhance stability and translational efficiency. The mRNA is encapsulated in lipid nanoparticles (LNPs) for effective delivery into host cells, facilitating the expression of the encoded antigen and the subsequent immune response [34].
Despite the straightforward nature of mRNA production compared to traditional vaccine methods, several manufacturing and quality control challenges persist. Key issues include the optimization of the production process to ensure high yields and purity of mRNA, as well as the stability of the final product. The purification steps often involve techniques such as chromatography and tangential flow filtration to remove contaminants and ensure the quality of the mRNA [7].
Regulatory approval for mRNA vaccines requires adherence to stringent guidelines that assess the quality, safety, and efficacy of the vaccines. Regulatory bodies, including the World Health Organization (WHO) and various national authorities, have been working towards establishing comprehensive guidelines to facilitate the development and evaluation of mRNA vaccines. This includes standardizing manufacturing practices and ensuring that vaccines meet the necessary quality benchmarks [35].
Furthermore, the rapid development of mRNA vaccines during the COVID-19 pandemic has highlighted the need for robust regulatory frameworks that can accommodate the fast-paced nature of vaccine innovation while ensuring public safety. Issues such as the need for proper dosage forms, the formulation of lipid nanoparticles, and the long-term stability of mRNA products are critical areas of focus [6].
The challenges faced in the manufacturing of mRNA vaccines are not limited to the production process alone. The complexities of scaling up production to meet global demand, ensuring the availability of high-quality raw materials, and managing the logistics of distribution also play significant roles in the overall success of mRNA vaccine deployment [32].
In conclusion, the development of mRNA vaccines involves intricate processes of design, manufacturing, and regulatory compliance. Addressing the associated challenges in quality control and manufacturing will be essential for the successful integration of mRNA technology into future vaccine strategies and for responding to emerging infectious diseases and public health crises.
6 Future Perspectives and Applications
6.1 Potential for Other Infectious Diseases
The development of mRNA vaccines represents a significant advancement in vaccinology, characterized by their ability to rapidly respond to emerging infectious diseases. The foundational technology behind mRNA vaccines involves the use of synthetic messenger RNA that encodes specific antigens from pathogens. This mRNA is delivered into host cells, where it directs the production of the encoded proteins, subsequently triggering an immune response.
The process of mRNA vaccine development begins with the identification of the target pathogen and the selection of appropriate antigens. These antigens are crucial as they are responsible for eliciting the desired immune response. Once the antigens are identified, the corresponding mRNA sequences are synthesized in vitro. This synthesis can be done using various techniques, including the use of linear DNA templates for transcription into mRNA, ensuring that the resultant mRNA is both functional and stable [10].
Delivery systems play a critical role in the effectiveness of mRNA vaccines. Lipid nanoparticles (LNPs) have emerged as the primary method for delivering mRNA into cells, protecting the mRNA from degradation and facilitating its uptake by target cells [36]. The design of these delivery systems is continually evolving to enhance stability, improve cellular uptake, and reduce potential adverse effects [37].
Recent advancements have shown that mRNA vaccines can be developed not only for viral infections, as evidenced by the rapid deployment of COVID-19 vaccines, but also for other infectious diseases, including bacterial and parasitic infections [38]. The versatility of mRNA technology allows for modifications that can address the specific challenges posed by different pathogens, including their mutation rates and immune evasion strategies [39].
Looking to the future, the potential applications of mRNA vaccines extend beyond infectious diseases to include cancer immunotherapy and the treatment of various other conditions. This is facilitated by the adaptability of mRNA platforms, which can be rapidly modified to encode different antigens as needed. Moreover, the ongoing research aims to optimize the immunogenicity of mRNA vaccines, enhance delivery mechanisms, and ensure global accessibility, particularly in resource-limited settings [1].
In conclusion, the development of mRNA vaccines is a dynamic and rapidly evolving field that holds promise for addressing a wide range of infectious diseases. The combination of swift design capabilities, potent immune responses, and scalable production methods positions mRNA vaccines as a transformative technology in the landscape of modern medicine. Future efforts will likely focus on overcoming existing challenges and expanding the applications of mRNA technology to combat both current and emerging health threats [3][37][40].
6.2 Therapeutic Applications Beyond Vaccines
mRNA vaccines are developed through a multi-step process that leverages advances in molecular biology and nanotechnology. The development begins with the design of the mRNA sequence, which encodes a specific antigen intended to elicit an immune response. This sequence is synthesized in vitro using a process called in vitro transcription (IVT), where a DNA template is transcribed into mRNA. The resulting mRNA is then encapsulated in lipid nanoparticles (LNPs) to protect it from degradation and facilitate its delivery into host cells [1][41].
The versatility of mRNA technology allows for rapid modifications to the antigen design, enabling quick responses to emerging infectious diseases and adaptations to viral mutations. This adaptability is crucial, especially in the context of infectious diseases, where pathogens can evolve rapidly [2][42]. Additionally, mRNA vaccines can be engineered to produce not only prophylactic effects but also therapeutic responses, targeting diseases such as cancer [43][44].
One of the key challenges in mRNA vaccine development is ensuring efficient delivery to the target cells. Various delivery systems are being explored, with lipid nanoparticles being the most widely used due to their favorable safety profile and efficacy in facilitating cellular uptake [36][41]. Moreover, ongoing research is focused on improving the stability of mRNA, enhancing its immunogenicity, and developing novel delivery vectors [45][46].
In terms of therapeutic applications beyond vaccines, mRNA technology is being explored for a range of treatments. For instance, mRNA-based therapies have shown promise in protein replacement therapy, cancer immunotherapy, and the treatment of genetic disorders. The ability of mRNA to instruct cells to produce therapeutic proteins directly in the body opens new avenues for treating conditions that currently lack effective therapies [38][47].
Looking forward, the future of mRNA technology appears promising. The development of self-amplifying mRNA and circular mRNA platforms is being investigated to enhance the expression of antigens and improve immune responses [2][47]. Furthermore, there is a growing interest in optimizing the design and delivery systems to overcome existing challenges, such as mRNA stability and the potential for off-target effects [10][43].
Overall, the ongoing advancements in mRNA vaccine development not only highlight their potential as a robust platform for infectious disease prevention but also pave the way for innovative therapeutic applications across various medical fields. As research continues to evolve, mRNA technology is poised to play a significant role in shaping the future of medicine [41][48].
7 Conclusion
The development of mRNA vaccines has revolutionized the field of vaccinology, demonstrating unprecedented speed and efficacy in response to health crises such as the COVID-19 pandemic. Key findings from this review highlight the significant advancements in mRNA technology, from its historical origins to its current applications. The integration of innovative design and synthesis methods, particularly the use of lipid nanoparticles for delivery, has enabled robust immune responses. Regulatory pathways have adapted to accommodate the rapid development of these vaccines, though challenges in manufacturing and quality control remain critical. Future research should focus on optimizing mRNA vaccine formulations, enhancing delivery systems, and exploring applications beyond infectious diseases, such as cancer therapy. The potential of mRNA technology is vast, and its continued evolution will be essential in addressing both current and emerging global health challenges.
References
- [1] Kai Yuan Leong;Seng Kong Tham;Chit Laa Poh. Revolutionizing immunization: a comprehensive review of mRNA vaccine technology and applications.. Virology journal(IF=3.8). 2025. PMID:40075519. DOI: 10.1186/s12985-025-02645-6.
- [2] Sha Li;Lu Zheng;Jingyi Zhong;Xihui Gao. Advancing mRNA vaccines for infectious diseases: key components, innovations, and clinical progress.. Essays in biochemistry(IF=5.7). 2025. PMID:40321006. DOI: 10.1042/EBC20253009.
- [3] Laura Matarazzo;Paulo J G Bettencourt. mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV.. Frontiers in immunology(IF=5.9). 2023. PMID:37168860. DOI: 10.3389/fimmu.2023.1172691.
- [4] Linlin Zheng;Han Feng. Respiratory virus mRNA vaccines: mRNA Design, clinical studies, and future challenges.. Animal models and experimental medicine(IF=3.4). 2025. PMID:40469015. DOI: 10.1002/ame2.70018.
- [5] Yalan Liu;Yuncheng Li;Qinxue Hu. Advances in saRNA Vaccine Research against Emerging/Re-Emerging Viruses.. Vaccines(IF=3.4). 2023. PMID:37514957. DOI: 10.3390/vaccines11071142.
- [6] Ivana Knezevic;Margaret A Liu;Keith Peden;Tiequn Zhou;Hye-Na Kang. Development of mRNA Vaccines: Scientific and Regulatory Issues.. Vaccines(IF=3.4). 2021. PMID:33498787. DOI: 10.3390/vaccines9020081.
- [7] Sara Sousa Rosa;Duarte M F Prazeres;Ana M Azevedo;Marco P C Marques. mRNA vaccines manufacturing: Challenges and bottlenecks.. Vaccine(IF=3.5). 2021. PMID:33771389. DOI: 10.1016/j.vaccine.2021.03.038.
- [8] Chaoying Hu;Yu Bai;Jianyang Liu;Yiping Wang;Qian He;Xuanxuan Zhang;Feiran Cheng;Miao Xu;Qunying Mao;Zhenglun Liang. Research progress on the quality control of mRNA vaccines.. Expert review of vaccines(IF=4.8). 2024. PMID:38733272. DOI: 10.1080/14760584.2024.2354251.
- [9] Mohamad Irfan Mohamad Razif;Nabilah Nizar;Nur Hannah Zainal Abidin;Syasya Nasuha Muhammad Ali;Wan Nurul Najihah Wan Zarimi;Junaidi Khotib;Deny Susanti;Muhammad Taufiq Mohd Jailani;Muhammad Taher. Emergence of mRNA vaccines in the management of cancer.. Expert review of vaccines(IF=4.8). 2023. PMID:37401128. DOI: 10.1080/14760584.2023.2232450.
- [10] Zhimeng Wei;Shuai Zhang;Xingya Wang;Ying Xue;Sheng Dang;Jingbo Zhai. Technological breakthroughs and advancements in the application of mRNA vaccines: a comprehensive exploration and future prospects.. Frontiers in immunology(IF=5.9). 2025. PMID:40103818. DOI: 10.3389/fimmu.2025.1524317.
- [11] Vrinda Gote;Pradeep Kumar Bolla;Nagavendra Kommineni;Arun Butreddy;Pavan Kumar Nukala;Sushesh Srivatsa Palakurthi;Wahid Khan. A Comprehensive Review of mRNA Vaccines.. International journal of molecular sciences(IF=4.9). 2023. PMID:36769023. DOI: 10.3390/ijms24032700.
- [12] Giulietta Maruggi;Cuiling Zhang;Junwei Li;Jeffrey B Ulmer;Dong Yu. mRNA as a Transformative Technology for Vaccine Development to Control Infectious Diseases.. Molecular therapy : the journal of the American Society of Gene Therapy(IF=12.0). 2019. PMID:30803823. DOI: 10.1016/j.ymthe.2019.01.020.
- [13] Jae-Hyeong Park;Kye Hun Kim. COVID-19 Vaccination-Related Myocarditis: What We Learned From Our Experience and What We Need to Do in The Future.. Korean circulation journal(IF=3.1). 2024. PMID:38654456. DOI: 10.4070/kcj.2024.0065.
- [14] John H Skerritt;Carolyn Tucek-Szabo;Brett Sutton;Terry Nolan. The Platform Technology Approach to mRNA Product Development and Regulation.. Vaccines(IF=3.4). 2024. PMID:38793779. DOI: 10.3390/vaccines12050528.
- [15] Abid Hussain;Maoye Wang;Dan Yu;Jiahui Zhang;Qais Ahmad Naseer;Aftab Ullah;Julien Milon Essola;Xu Zhang. Medical and molecular biophysical techniques as substantial tools in the era of mRNA-based vaccine technology.. Biomaterials science(IF=5.7). 2024. PMID:39016519. DOI: 10.1039/d4bm00561a.
- [16] Naveen Kumar;Nicholas C Hazell;Jiani Bei;Tina Nguyen;Haitao Hu. PCR-generated DNA templates enable efficient, rapid, and cost-effective mRNA synthesis.. Methods (San Diego, Calif.)(IF=4.3). 2025. PMID:40825451. DOI: 10.1016/j.ymeth.2025.08.007.
- [17] Alina Hengelbrock;Axel Schmidt;Jochen Strube. Digital Twin Fundamentals of mRNA In Vitro Transcription in Variable Scale Toward Autonomous Operation.. ACS omega(IF=4.3). 2024. PMID:38405539. DOI: 10.1021/acsomega.3c08732.
- [18] Sara Sousa Rosa;Davide Nunes;Luis Antunes;Duarte M F Prazeres;Marco P C Marques;Ana M Azevedo. Maximizing mRNA vaccine production with Bayesian optimization.. Biotechnology and bioengineering(IF=3.6). 2022. PMID:36017534. DOI: 10.1002/bit.28216.
- [19] Emma N Welbourne;Kate A Loveday;Adithya Nair;Ehsan Nourafkan;Jixin Qu;Ken Cook;Zoltán Kis;Mark J Dickman. Anion exchange HPLC monitoring of mRNA in vitro transcription reactions to support mRNA manufacturing process development.. Frontiers in molecular biosciences(IF=4.0). 2024. PMID:38516194. DOI: 10.3389/fmolb.2024.1250833.
- [20] Benoît Levast;Sandra Schulz;Sylvia van Drunen Littel-van den Hurk;Volker Gerdts. Animal models for neonatal diseases in humans.. Vaccine(IF=3.5). 2013. PMID:23238328. DOI: .
- [21] Volker Gerdts;Heather L Wilson;Francois Meurens;Sylvia van Drunen Littel-van den Hurk;Don Wilson;Stewart Walker;Colette Wheler;Hugh Townsend;Andrew A Potter. Large animal models for vaccine development and testing.. ILAR journal(IF=3.1). 2015. PMID:25991698. DOI: 10.1093/ilar/ilv009.
- [22] Tjessa Bondue;Sante Princiero Berlingerio;Lambertus van den Heuvel;Elena Levtchenko. The Zebrafish Embryo as a Model Organism for Testing mRNA-Based Therapeutics.. International journal of molecular sciences(IF=4.9). 2023. PMID:37446400. DOI: 10.3390/ijms241311224.
- [23] Yihan Lu;Ciying Qian;Yang Huang;Tianyu Ren;Wuzhi Xie;Ningshao Xia;Shaowei Li. Advancing mRNA vaccines: A comprehensive review of design, delivery, and efficacy in infectious diseases.. International journal of biological macromolecules(IF=8.5). 2025. PMID:40571008. DOI: 10.1016/j.ijbiomac.2025.145501.
- [24] Winthrop Kl. mRNA vaccines in the rheumatologist's future.. Seminars in arthritis and rheumatism(IF=4.4). 2024. PMID:38040517. DOI: 10.1016/j.semarthrit.2023.152325.
- [25] Sola Oloruntimehin;Florence Akinyi;Michael Paul;Olumuyiwa Ariyo. mRNA Vaccine Technology Beyond COVID-19.. Vaccines(IF=3.4). 2025. PMID:40573932. DOI: 10.3390/vaccines13060601.
- [26] Jonathan L Golob;Njira Lugogo;Adam S Lauring;Anna S Lok. SARS-CoV-2 vaccines: a triumph of science and collaboration.. JCI insight(IF=6.1). 2021. PMID:33822773. DOI: .
- [27] Courtney L Finch;Christian Martinez;Elizabeth Leffel;Mario H Skiadopoulos;Adam Hacker;Betty Mwesigwa;Diadié Maïga;Ian Mugisa;Grant Munkwase;Roxana Rustomjee. Vaccine Licensure in the Absence of Human Efficacy Data.. Vaccines(IF=3.4). 2022. PMID:35335000. DOI: 10.3390/vaccines10030368.
- [28] Rossella Brandi;Alessia Paganelli;Raffaele D'Amelio;Paolo Giuliani;Florigio Lista;Simonetta Salemi;Roberto Paganelli. mRNA Vaccines Against COVID-19 as Trailblazers for Other Human Infectious Diseases.. Vaccines(IF=3.4). 2024. PMID:39772079. DOI: 10.3390/vaccines12121418.
- [29] Jennifer A Whitaker;Hana M El Sahly;C Mary Healy. mRNA vaccines against respiratory viruses.. Current opinion in infectious diseases(IF=4.0). 2023. PMID:37462930. DOI: 10.1097/QCO.0000000000000948.
- [30] Athina Sofroniou;Anna Ridley. A Systematic Review of Clinical Trials Using mRNA Vaccines for Infectious Diseases other than COVID-19.. British journal of biomedical science(IF=4.6). 2025. PMID:40756606. DOI: 10.3389/bjbs.2025.14557.
- [31] Alexey V Yaremenko;Muhammad Muzamil Khan;Xueyan Zhen;Yan Tang;Wei Tao. Clinical advances of mRNA vaccines for cancer immunotherapy.. Med (New York, N.Y.)(IF=11.8). 2025. PMID:39798545. DOI: 10.1016/j.medj.2024.11.015.
- [32] Maryam Youssef;Cynthia Hitti;Julia Puppin Chaves Fulber;Amine A Kamen. Enabling mRNA Therapeutics: Current Landscape and Challenges in Manufacturing.. Biomolecules(IF=4.8). 2023. PMID:37892179. DOI: 10.3390/biom13101497.
- [33] Shyam Rele. COVID-19 vaccine development during pandemic: gap analysis, opportunities, and impact on future emerging infectious disease development strategies.. Human vaccines & immunotherapeutics(IF=3.5). 2021. PMID:32993453. DOI: 10.1080/21645515.2020.1822136.
- [34] Saeideh Khorshid Sokhangouy;Matine Behzadi;Shokuh Rezaei;Mahsa Farjami;Maryam Haghshenas;Yahya Sefidbakht;Sina Mozaffari-Jovin. mRNA Vaccines: Design Principles, Mechanisms, and Manufacturing-Insights From COVID-19 as a Model for Combating Infectious Diseases.. Biotechnology journal(IF=3.1). 2025. PMID:39989260. DOI: 10.1002/biot.202400596.
- [35] Margaret A Liu;Tiequn Zhou;Rebecca L Sheets;Heidi Meyer;Ivana Knezevic. WHO informal consultation on regulatory considerations for evaluation of the quality, safety and efficacy of RNA-based prophylactic vaccines for infectious diseases, 20-22 April 2021.. Emerging microbes & infections(IF=7.5). 2022. PMID:35001848. DOI: 10.1080/22221751.2022.2026742.
- [36] Basmah N Aldosari;Iman M Alfagih;Alanood S Almurshedi. Lipid Nanoparticles as Delivery Systems for RNA-Based Vaccines.. Pharmaceutics(IF=5.5). 2021. PMID:33540942. DOI: 10.3390/pharmaceutics13020206.
- [37] Dariush Haghmorad;Majid Eslami;Niloufar Orooji;Iryna Halabitska;Iryna Kamyshna;Oleksandr Kamyshnyi;Valentyn Oksenych. mRNA vaccine platforms: linking infectious disease prevention and cancer immunotherapy.. Frontiers in bioengineering and biotechnology(IF=4.8). 2025. PMID:40144393. DOI: 10.3389/fbioe.2025.1547025.
- [38] Nouran Rezk;Siobhán McClean. Harnessing the Potential of mRNA Vaccines Against Infectious Diseases.. Microbial biotechnology(IF=5.2). 2025. PMID:40853332. DOI: 10.1111/1751-7915.70212.
- [39] Sora Son;Kyuri Lee. Development of mRNA Vaccines/Therapeutics and Their Delivery System.. Molecules and cells(IF=6.5). 2023. PMID:36697236. DOI: 10.14348/molcells.2023.2165.
- [40] Zhen Zhang;Jingli Du;Danyang Zhang;Rui Han;Xueqiong Wu;Yan Liang. Research progress of mRNA vaccines for infectious diseases.. European journal of medical research(IF=3.4). 2025. PMID:40847375. DOI: 10.1186/s40001-025-03060-x.
- [41] Nojoud Al Fayez;Majed S Nassar;Abdullah A Alshehri;Meshal K Alnefaie;Fahad A Almughem;Bayan Y Alshehri;Abdullah O Alawad;Essam A Tawfik. Recent Advancement in mRNA Vaccine Development and Applications.. Pharmaceutics(IF=5.5). 2023. PMID:37514158. DOI: 10.3390/pharmaceutics15071972.
- [42] Zhongyan Wu;Weilu Sun;Hailong Qi. Recent Advancements in mRNA Vaccines: From Target Selection to Delivery Systems.. Vaccines(IF=3.4). 2024. PMID:39203999. DOI: 10.3390/vaccines12080873.
- [43] Londiwe Simphiwe Mbatha;Jude Akinyelu;Fiona Maiyo;Tukayi Kudanga. Future prospects in mRNA vaccine development.. Biomedical materials (Bristol, England)(IF=3.7). 2023. PMID:37589309. DOI: 10.1088/1748-605X/aceceb.
- [44] Mia Karam;Georges Daoud. mRNA vaccines: Past, present, future.. Asian journal of pharmaceutical sciences(IF=11.9). 2022. PMID:36105317. DOI: 10.1016/j.ajps.2022.05.003.
- [45] Qiannan Cao;Huapan Fang;Huayu Tian. mRNA vaccines contribute to innate and adaptive immunity to enhance immune response in vivo.. Biomaterials(IF=12.9). 2024. PMID:38820767. DOI: 10.1016/j.biomaterials.2024.122628.
- [46] Mingyue Pan;Weiling Cao;Jingbo Zhai;Chunfu Zheng;Yingying Xu;Peng Zhang. mRNA-based vaccines and therapies - a revolutionary approach for conquering fast-spreading infections and other clinical applications: a review.. International journal of biological macromolecules(IF=8.5). 2025. PMID:40233916. DOI: 10.1016/j.ijbiomac.2025.143134.
- [47] Huan-Huan Wei;Liangliang Zheng;Zefeng Wang. mRNA therapeutics: New vaccination and beyond.. Fundamental research(IF=6.3). 2023. PMID:38933291. DOI: 10.1016/j.fmre.2023.02.022.
- [48] Cristiana Perrotta;Claudio Fenizia;Carla Carnovale;Marco Pozzi;Daria Trabattoni;Davide Cervia;Emilio Clementi. Updated Considerations for the Immunopharmacological Aspects of the "Talented mRNA Vaccines".. Vaccines(IF=3.4). 2023. PMID:37766157. DOI: 10.3390/vaccines11091481.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
mRNA vaccines · vaccine technology · immune response · clinical development · regulatory pathways
© 2025 MaltSci
