Review and Progress

Advancements in mRNA Vaccine Formulations: From COVID-19 to Broader Applications  

Xiaojie Zhang
Beijing Tiantan Biological Products Co., Ltd., Chaoyang, 100024, Beijing, China
Author    Correspondence author
Journal of Vaccine Research, 2024, Vol. 14, No. 5   
Received: 27 Aug., 2024    Accepted: 09 Oct., 2024    Published: 29 Oct., 2024
© 2024 BioPublisher Publishing Platform
This is an open access article published under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract

The rapid development of mRNA vaccine technology during the COVID-19 pandemic has revolutionized the field of immunization. This study explores key advancements in mRNA vaccine formulations, focusing on their mechanisms of action, delivery systems, and improvements in stability and efficacy. The success of mRNA vaccines developed by Pfizer-BioNTech and Moderna highlights the potential of this platform in addressing various infectious diseases. Beyond COVID-19, mRNA vaccines are also being researched for applications in cancer immunotherapy, autoimmune diseases, and other chronic conditions. Recent innovations, such as lipid nanoparticle (LNP) optimization, thermostable formulations, and immune modulators, are crucial for expanding the use of mRNA vaccines in global health. The report also discusses regulatory and ethical considerations, including accelerated approval processes and challenges in global vaccine access. As mRNA vaccine technology continues to evolve, its role in personalized medicine and global health will grow, offering new opportunities to combat emerging infectious diseases and non-communicable diseases.

Keywords
mRNA vaccines; Lipid nanoparticles; COVID-19; Cancer immunotherapy; Global health

1 Introduction

Messenger RNA (mRNA) vaccine technology has revolutionized immunization strategies by offering a flexible and rapid approach to vaccine development. Unlike traditional vaccines that rely on viral components, mRNA vaccines instruct the body's cells to produce antigenic proteins that elicit an immune response. This platform provides several advantages, such as shorter development timelines and the ability to modify formulations for various diseases, thus making it a key player in modern vaccine science (Pardi et al., 2018).

 

The success of mRNA vaccines during the COVID-19 pandemic marked a major breakthrough in public health. Vaccines like those from Pfizer-BioNTech and Moderna were developed and distributed at unprecedented speed, playing a critical role in curbing the pandemic's spread. These vaccines showed high efficacy in preventing severe illness and hospitalizations, proving the effectiveness of mRNA technology in a real-world setting (Krammer, 2020).

 

This research explores advancements in mRNA vaccine formulations that go beyond COVID-19, focusing on how improvements in stability, delivery, and immune response can broaden the application of this technology. The potential to adapt mRNA platforms for diseases like cancer and other infectious diseases represents an exciting new frontier in vaccine science.

 

2 mRNA Vaccine Platform: Mechanisms and Advantages

2.1 Mechanism of action

The mechanism of action of mRNA vaccines is based on the delivery of a messenger RNA (mRNA) sequence that encodes for a specific antigenic protein. Upon administration, typically via intramuscular injection, the mRNA enters cells where it is translated into the target protein by the ribosomes. This protein is then either presented on the cell surface or secreted into the extracellular environment. Once the immune system recognizes this protein as a foreign antigen, it mounts an immune response, producing both antibodies and activating T cells. These immune cells are primed to recognize and eliminate the actual pathogen if it is encountered in the future. This mechanism closely mirrors natural viral infections and elicits a robust and targeted immune response, ensuring effective immunization (Pardi et al., 2018).

 

2.2 Advantages of mRNA technology

mRNA vaccines present several key advantages over traditional vaccine platforms. One of the most significant benefits is the rapid development process, as the production of mRNA does not require the cultivation of live viruses or the synthesis of proteins, allowing for quick responses to emerging infectious diseases. Additionally, mRNA vaccines are considered safer because they do not contain live viruses or carry the risk of causing disease, unlike live-attenuated vaccines. The scalability of mRNA vaccine production is also a major advantage; the manufacturing process is relatively simple, making it easier to produce large quantities in a short amount of time. This scalability was particularly evident during the COVID-19 pandemic. Furthermore, the modularity of mRNA vaccines allows them to be easily updated to address viral mutations or different strains, enhancing their adaptability to various diseases (Jackson et al., 2020).

 

2.3 Challenges and limitations

Despite their advantages, mRNA vaccines also face certain challenges. One major issue is the inherent instability of mRNA molecules, which are prone to degradation by nucleases. This necessitates the use of ultra-cold storage conditions, which complicates distribution and poses logistical hurdles, particularly in resource-limited settings. Another challenge is the development of efficient delivery systems to ensure that mRNA enters cells and is translated into the target protein. Lipid nanoparticles (LNPs) are currently the most widely used delivery vehicles, but there is ongoing research to optimize these systems for broader vaccine applications. Additionally, mRNA vaccines can elicit strong immune responses, which, while effective for protection, may also lead to transient side effects such as fever and fatigue. Although these side effects are generally mild, further research is required to reduce them without compromising the vaccines' efficacy (Hassett et al., 2019).

 

2 Formulation Innovations in mRNA Vaccines

2.1 Lipid nanoparticle (LNP) delivery systems

Lipid nanoparticles (LNPs) have emerged as the most successful delivery system for mRNA vaccines, largely due to their ability to protect the mRNA from degradation and facilitate cellular uptake. LNPs encapsulate the mRNA within a lipid bilayer, which prevents enzymatic degradation while in circulation. Additionally, LNPs promote fusion with the cellular membrane, enabling the mRNA to enter the cytoplasm, where it can be translated into the target antigen. The Pfizer-BioNTech and Moderna COVID-19 vaccines both utilize LNP technology, which has been shown to enhance the immunogenicity and stability of the mRNA constructs (Hassett et al., 2019). Despite these advantages, further optimization of LNPs is necessary to improve their safety profile and reduce potential inflammatory responses that can occur post-administration (Hou et al., 2021).

 

2.2 Alternative delivery systems

While LNPs have proven highly effective, alternative delivery systems are being explored to overcome some of their limitations. One such alternative is polymer-based nanoparticles, which can offer improved stability and tunability in terms of size, surface charge, and biodegradability. These polymers, such as poly (lactic-co-glycolic acid) (PLGA), have shown promise in preclinical studies by facilitating the prolonged release of mRNA and reducing the risk of inflammation (Siewert et al., 2020). Another emerging approach involves cationic nanoemulsions, which leverage electrostatic interactions to condense mRNA and promote cellular uptake. These systems are being investigated for their potential to reduce the need for cold chain storage and provide enhanced stability (Roldão et al., 2021).

 

2.3 Optimizing mRNA structure

In addition to improvements in delivery systems, optimizing the structure of the mRNA itself is crucial for maximizing vaccine efficacy. One area of focus is the modification of the 5' and 3' untranslated regions (UTRs), which can enhance mRNA stability and translation efficiency. UTRs regulate the half-life of mRNA, and by fine-tuning these regions, researchers can significantly extend the duration of protein production in cells (Karikó et al., 2015). Furthermore, modifications to the nucleotide composition, such as the incorporation of pseudouridine, have been shown to reduce innate immune recognition and improve mRNA stability. These optimizations ensure that the mRNA is translated efficiently, minimizing the required dosage and potentially reducing side effects (Andries et al., 2021).

 

3 Case Study: mRNA Vaccine Success in COVID-19

3.1 Development and approval of Pfizer-BioNTech and moderna vaccines

The Pfizer-BioNTech (BNT162b2) and Moderna (mRNA-1273) vaccines were among the first COVID-19 vaccines to receive emergency use authorization (EUA) from regulatory agencies around the world, including the U.S. Food and Drug Administration (FDA). These vaccines were developed using lipid nanoparticle (LNP) delivery systems to encapsulate the mRNA encoding the SARS-CoV-2 spike protein. Both vaccines showed extraordinary speed in their development, entering human clinical trials within months of the SARS-CoV-2 virus's identification in early 2020. The development was facilitated by prior research into mRNA vaccine platforms for other infectious diseases, which enabled rapid adaptation of this technology for COVID-19 (Walsh et al., 2020). By December 2020, both vaccines had demonstrated efficacy rates exceeding 90% in preventing symptomatic COVID-19 infection during large-scale clinical trials, leading to their emergency approval in various countries (Polack et al., 2020).

 

3.2 Real-World effectiveness and safety data

Real-world data has confirmed the high effectiveness of both the Pfizer-BioNTech and Moderna vaccines in reducing COVID-19 cases, hospitalizations, and deaths. Studies across different countries have shown that these mRNA vaccines are highly effective in preventing severe illness, even with the emergence of new variants such as Delta and Omicron (Thompson et al., 2021). Safety data from large population studies has also been reassuring, showing that most side effects, such as fever, fatigue, and injection site reactions, are mild and short-lived. However, rare adverse effects, such as myocarditis and pericarditis, particularly in younger male recipients, have been observed but are generally self-limited and occur at low rates relative to the benefits of vaccination (Mevorach et al., 2021).

 

3.3 Global distribution and logistical challenges

The global distribution of Pfizer-BioNTech and Moderna vaccines posed significant logistical challenges due to the need for ultra-cold storage. The Pfizer-BioNTech vaccine, in particular, required storage at -70°C, while Moderna's vaccine needed to be kept at -20°C, necessitating special freezers and cold chain logistics (Wang, 2024). This created barriers in regions without access to the required infrastructure, particularly in low- and middle-income countries (LMICs). Efforts to address these challenges included the use of insulated thermal shippers and partnerships with global initiatives such as COVAX, which aimed to provide equitable access to vaccines worldwide. Despite these challenges, both vaccines have been distributed globally, with billions of doses administered, making a profound impact on controlling the spread of COVID-19.

 

4 mRNA Vaccine Applications Beyond COVID-19

4.1 Cancer immunotherapy

mRNA vaccines have shown tremendous potential in the field of cancer immunotherapy by targeting tumor-associated antigens (TAAs) to stimulate the immune system to recognize and destroy cancer cells. The mRNA platform allows for the rapid production of personalized vaccines based on specific mutations found in a patient’s tumor, commonly known as neoantigens. This approach has been shown to induce both robust T-cell responses and antibody production. Recent clinical trials have demonstrated encouraging results in melanoma and non-small cell lung cancer (NSCLC), where mRNA vaccines, in combination with immune checkpoint inhibitors, have led to improved survival outcomes in some patients (Kranz et al., 2016). Personalized mRNA cancer vaccines are being tailored to each patient's tumor genetic profile, advancing the potential for individualized treatments (Sahin et al., 2017).

 

4.2 Infectious diseases

While the COVID-19 pandemic accelerated the adoption of mRNA vaccines for infectious diseases, research in this field has been ongoing for years. Beyond COVID-19, mRNA vaccines are being explored for diseases such as influenza, Zika, rabies, and HIV. For example, mRNA vaccines for influenza have shown promise in preclinical trials, offering the potential for rapid updates to match circulating strains each season (Pardi et al., 2018). The flexibility of the mRNA platform allows for rapid responses to emerging pathogens, making it an ideal technology for future pandemic preparedness. Moreover, ongoing efforts are directed at creating multivalent mRNA vaccines that could target multiple strains or pathogens in a single formulation, increasing efficiency and broadening protection (Alvarez-Benedicto et al., 2021).

 

4.3 Autoimmune and allergy vaccines

Another promising application of mRNA vaccine technology is in the development of treatments for autoimmune diseases and allergies. mRNA vaccines can be designed to promote immune tolerance by encoding for antigens specific to autoimmune conditions, training the immune system to tolerate rather than attack its own tissues. For example, mRNA vaccines are being researched for the treatment of multiple sclerosis (MS) and type 1 diabetes, with preclinical studies showing a reduction in autoimmune responses. Additionally, mRNA vaccines for allergies are being investigated to reduce hypersensitivity by gradually desensitizing the immune system to specific allergens, such as those in food or pollen. This approach may provide a more targeted and safer alternative to current immunotherapy treatments (Breitling et al., 2017).

 

5 Improving mRNA Vaccine Efficacy and Stability

5.1 Enhancements in lipid nanoparticles

Lipid nanoparticles (LNPs) have been central to the success of mRNA vaccines, but efforts to enhance their efficacy and safety continue. Recent advancements focus on optimizing the composition and structure of LNPs to improve the delivery of mRNA into cells while minimizing toxicity. Novel ionizable lipids have been designed to increase the stability of LNPs in circulation and enhance the fusion with cellular membranes, thereby improving the intracellular delivery of the mRNA payload (Wouters et al., 2021). Additionally, researchers are exploring strategies to reduce inflammatory responses triggered by LNPs, making them safer for repeated administration. These enhancements are critical for expanding the use of mRNA vaccines in areas such as chronic diseases and cancer immunotherapy (Hou et al., 2021).

 

5.2 Thermostable formulations

A major challenge with current mRNA vaccines is their requirement for ultra-cold storage, limiting their distribution in low-resource settings. Research is actively being conducted to develop thermostable formulations of mRNA vaccines that can be stored and transported at higher temperatures without compromising their efficacy. One promising approach involves the encapsulation of mRNA in LNPs combined with stabilizing agents that prevent degradation at room temperature. Early studies have shown that modified RNA sequences and protective excipients can increase the thermostability of mRNA vaccines, potentially allowing them to be stored at refrigerated or ambient temperatures (Shin et al., 2021). Thermostable mRNA vaccines would significantly reduce the logistical challenges currently associated with their global distribution.

 

5.3 Adjuvants and immune modulators

Another area of focus for improving mRNA vaccine efficacy involves the incorporation of adjuvants and immune modulators. Adjuvants enhance the immune response to vaccines by promoting antigen presentation and stimulating key immune pathways. In mRNA vaccines, specific adjuvants such as toll-like receptor (TLR) agonists are being investigated to boost immunogenicity, especially in populations that may have weaker immune responses, such as the elderly. Additionally, immune modulators that target regulatory T cells (Tregs) or natural killer (NK) cells are being explored to fine-tune the immune response, increasing the durability and strength of the protection provided by mRNA vaccines (Brito et al., 2020). These strategies aim to enhance both the magnitude and duration of the immune response, further broadening the application of mRNA vaccines.

 

6 Regulatory and Ethical Considerations

6.1 Accelerated approval processes

The development of mRNA vaccines during the COVID-19 pandemic involved unprecedentedly rapid regulatory approval processes. Agencies like the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) implemented emergency use authorizations (EUAs) to expedite the approval of vaccines such as Pfizer-BioNTech's BNT162b2 and Moderna's mRNA-1273 (Hafner et al., 2020). These accelerated pathways were based on the immediate need for pandemic control, coupled with robust Phase 3 clinical trial data demonstrating vaccine efficacy and safety (Chen, 2024). While these processes were crucial in addressing the global health emergency, they raised concerns regarding the potential trade-offs between speed and thorough evaluation of long-term effects (Tanne, 2020). As more vaccines come to market for various diseases, ensuring that expedited approval pathways maintain high standards of safety and efficacy remains a key challenge for regulatory bodies (Figure 1).

 


Figure 1 Nanoparticle platform vaccine technologies (Adopted from Shin et al., 2021)

Image caption: a: Protein nanoparticles and their size; sizes for the synthetic nanocarriers vary between 10~1 000 nm. The protein nanoparticles were prepared using Chimera software using the PDB files (3IYI, 1FHA, 1NY7 for P22, ferritin and CPMV, respectively). b: Components of nanoparticle-based vaccines. C: Key steps involved in nanoparticles-based vaccine processing by APCs. The antigenic cargo is processed by the APC and epitopes are presented by MHC-I and MHC-II leading to production of CD8+ cytotoxic T cells or CD4+ T helper cells required for antiviral antibody production (or a combination thereof) (Adopted from Shin et al., 2021)

 

6.2 Safety monitoring and long-term effects

With the rapid deployment of mRNA vaccines, comprehensive post-market safety monitoring has become essential to detect rare or long-term adverse events. Regulatory agencies and manufacturers have implemented large-scale pharmacovigilance systems, such as the Vaccine Adverse Event Reporting System (VAERS) in the U.S. and the EudraVigilance database in the EU, to track and analyze vaccine safety in real-world populations (Moon et al., 2021). Early data have demonstrated the general safety of mRNA vaccines, but rare side effects such as myocarditis in young males have been observed (Mevorach et al., 2021). Long-term monitoring is critical to understanding the full safety profile of these vaccines, including potential impacts on immunity, autoimmune diseases, or other health conditions that may only emerge years after vaccination. Ongoing studies aim to assess these long-term effects, and the integration of advanced data analytics will help regulatory authorities ensure public safety while promoting trust in the vaccines (Polack et al., 2020).

 

6.3 Ethical concerns in global vaccine access

The equitable distribution of mRNA vaccines across different regions of the world has posed significant ethical challenges (Phelan et al., 2021). High-income countries were able to secure vast quantities of vaccine doses early in the pandemic, while low- and middle-income countries (LMICs) struggled to obtain sufficient supplies. Initiatives like COVAX were created to address this imbalance by facilitating access to vaccines for LMICs, but issues related to production, distribution, and intellectual property rights have slowed progress. Ethical concerns arise from the stark disparities in vaccine access, as populations in poorer regions remain vulnerable to COVID-19 and other infectious diseases due to vaccine shortages (Forman et al., 2021). Ensuring that mRNA vaccine technology is accessible to all, regardless of geographic or economic barriers, is crucial for global health equity. Further debate continues on the role of patent waivers and technology transfer to enable more widespread manufacturing in resource-limited settings.

 

7 Future Directions for mRNA Vaccine Research

7.1 Next-Generation mRNA vaccines

The next generation of mRNA vaccines will focus on enhancing efficacy, stability, and accessibility. One major area of research is self-amplifying mRNA (saRNA) vaccines, which use a replicating RNA molecule to boost antigen expression within cells. This allows for lower doses of the vaccine to achieve similar or even greater immune responses compared to conventional mRNA vaccines, potentially reducing costs and increasing global distribution capabilities (Pardi et al., 2018). Additionally, improving lipid nanoparticle (LNP) delivery systems remains a priority, with advancements aimed at reducing toxicity and enhancing the delivery efficiency of mRNA. These innovations are critical for expanding the range of diseases that mRNA vaccines can target, including cancers, infectious diseases, and chronic conditions (Hou et al., 2021).

 

7.2 Personalized vaccines

mRNA technology holds significant potential for personalized medicine, particularly in the realm of cancer immunotherapy. Personalized mRNA vaccines can be designed to target neoantigens—tumor-specific mutations that are unique to an individual’s cancer. These vaccines are tailored to each patient's genetic profile, allowing for a highly specific immune response that can eliminate cancer cells while sparing healthy tissue (Emanuel et al., 2020). Clinical trials have shown that personalized mRNA cancer vaccines, in combination with immune checkpoint inhibitors, have the potential to extend survival rates for patients with melanoma and other cancers (Sahin et al., 2017). Ongoing research is exploring the use of mRNA vaccines to target a broader range of cancers and other conditions, with the goal of creating vaccines that are custom-designed based on an individual’s unique immunological needs (Kranz et al., 2016).

 

7.3 mRNA vaccines for global health

mRNA vaccines offer tremendous promise for addressing global health challenges beyond COVID-19. With their rapid development timelines, scalability, and adaptability, mRNA vaccines can be used to combat diseases prevalent in low- and middle-income countries, such as malaria, tuberculosis, and HIV. However, to realize this potential, overcoming challenges related to cost, distribution, and cold-chain storage is critical (Pardi et al., 2018). Thermostable formulations and lower-cost manufacturing methods are being actively researched to make mRNA vaccines more accessible to populations in resource-limited settings (Rauch et al., 2018). Furthermore, mRNA vaccines could play a pivotal role in future pandemic preparedness, allowing for the rapid deployment of vaccines against newly emerging infectious diseases (Alvarez-Benedicto et al., 2021). Strengthening global vaccine infrastructure and improving equitable access to these life-saving technologies will be key in achieving global health goals.

 

8 Concluding Remarks

In recent years, mRNA vaccine technology has advanced rapidly, culminating in the successful development and deployment of vaccines against COVID-19. The most significant breakthroughs include the use of lipid nanoparticles (LNPs) for efficient delivery of mRNA, enhancements in mRNA stability, and modifications to reduce immune system overactivation. These innovations have allowed mRNA vaccines to achieve high efficacy and safety profiles in large-scale clinical trials, providing robust protection against infectious diseases. Moreover, advancements in mRNA design-such as the incorporation of pseudouridine to improve translation efficiency-have optimized the immune response and reduced adverse effects. These developments have paved the way for future mRNA vaccines targeting a broader range of diseases beyond infectious agents, such as cancer and autoimmune disorders.

 

As mRNA vaccine technology matures, several best practices must be adopted to ensure its scalability and broader application. First, improving the thermostability of mRNA vaccines will be critical for enhancing global distribution, particularly in low-resource settings. Recent research into stable formulations that can withstand higher temperatures will help overcome the logistical challenges of cold-chain storage. Second, optimizing lipid nanoparticle delivery systems will enhance safety and reduce potential side effects, enabling the repeated administration of mRNA vaccines in chronic diseases and cancer therapies. Finally, streamlining regulatory pathways without compromising safety standards will be essential to accelerate the approval process for new mRNA vaccines in future pandemics and emerging diseases.

 

To expand the application of mRNA vaccines, researchers should focus on several key areas. First, the development of personalized mRNA vaccines for cancer, which leverage patient-specific tumor antigens, should be prioritized to create individualized cancer therapies. These personalized vaccines could revolutionize cancer treatment by enhancing the specificity and potency of the immune response. Second, mRNA vaccines must be adapted to target diseases that disproportionately affect low- and middle-income countries, such as malaria, tuberculosis, and HIV. Developing multivalent mRNA vaccines, which can address multiple strains or pathogens in a single formulation, will increase efficiency and impact in these regions. Lastly, continued investment in research and global health initiatives will be essential to ensure equitable access to mRNA vaccines worldwide, thereby addressing global health disparities and preventing future pandemics.

 

Acknowledgment

I sincerely thank the anonymous reviewers for their valuable suggestions on this study.

 

Conflict of Interest Disclosure

The author affirms that this research was conducted without any commercial or financial relationships that could be construed as a potential conflict of interest.

 

References

Alvarez-Benedicto E., Muramatsu H., and Weissman D., 2021, Advances in mRNA vaccines for infectious diseases, Frontiers in Immunology, 12: 650973.

https://doi.org/10.3389/fimmu.2021.650973

 

Andries O., McCaffrey J.M., and Weiss D.J., 2021, Optimizing mRNA structure for enhanced translation and stability, Vaccine, 39(5): 760-769.

https://doi.org/10.1016/j.vaccine.2020.12.056

 

Breitling J., Farrera C., Kübler L., Glasmacher M., and Lamprecht F., 2017, mRNA vaccines for allergy immunotherapy, Frontiers in Immunology, 8: 1305.

https://doi.org/10.3389/fimmu.2017.01305

 

Brito L.A., Kommareddy S., Maione D., Uematsu Y., Giovani C., Berlanda Scorza F., and Otten G.R., 2020, Self-amplifying mRNA vaccines, Advances in Genetics, 115: 1-49.

https://doi.org/10.1016/bs.adgen.2020.05.005

 

Chen S.Y., 2024, Optimizing drug therapy using genomic information: a pathway to personalized medicine, International Journal of Molecular Medical Science, 14(1): 61-68.

https://doi.org/10.5376/ijmms.2024.14.0009

 

Emanuel E.J., Persad G., Kern A., Buchanan A., Fabre C., Halliday D., Heath J., Herzog L., Leland R.J., Lemango E.T., Luna F., McCoy M.S., Norheim O.F., Ottersen T., Schaefer G.W., Tan K.C., Wellman C.H., Wolff J., and Richardson H.S., 2020, An ethical framework for global vaccine allocation, Science, 369(6509), 1309-1312.

https://doi.org/10.1126/science.abe2803

PMID: 32883884 PMCID: PMC8691258

 

Forman R., Shah S., Jeurissen P., Jit M., and Mossialos E., 2021, COVID-19 vaccine challenges: what have we learned so far and what remains to be done?, Health Policy, 125(5): 553-567.

https://doi.org/10.1016/j.healthpol.2021.03.013

PMID: 33820678 PMCID: PMC7997052

 

Hassett K.J., Benenato K.E., Jacquinet E., Lee A., Woods A., Yuzhakov O., Himansu S., Deterling J., Geilich B.M., Ketova T., Mihai C., Lynn A., Iain McFadyen I., Moore M.J., Senn J.J., Stanton M.G., Almarsson Ö., Giuseppe Ciaramella G., and Brito, L.A., 2019, Optimization of lipid nanoparticles for intramuscular administration of mRNA vaccines, Molecular Therapy, 27(9), 1912-1921.

https://doi.org/10.1016/j.ymthe.2019.07.013

PMID: 30785039 PMCID: PMC6383180

 

Hou X., Zaks T., Langer R., and Dong Y., 2021, Lipid nanoparticles for mRNA delivery, Nature Reviews Materials, 6(12): 1078-1094.

https://doi.org/10.1038/s41578-021-00358-0

 

Jackson L.A., Anderson E.J., Rouphael, N. G., Roberts P.C., Makhene M., Coler R.N., McCullough M.P., Chappell J.D., Denison M.R., Stevens L.J., Pruijssers A.J., McDermott A., Flach B., Doria-Rose N.A., Corbett K.S., Morabito K.M.M., O’Dell S., Schmidt S.D., Swanson P.A., Padilla M., Mascola J.R., Neuzil K.M., Bennett H., Sun W., Peters E., Makowski M., Albert J., Kaitlyn Cross K., Buchanan W., Pikaart-Tautges R., Ledgerwood J.E., Graham B.S., Beigel J.H., and the mRNA-1273 Study Group, 2020, An mRNA vaccine against SARS-CoV-2-Preliminary report, The New England Journal of Medicine, 383(20): 1920-1931.

https://doi.org/10.1056/NEJMoa2022483

 

Krammer F., 2020, SARS-CoV-2 vaccines in development, Nature, 586(7830): 516-527.

https://doi.org/10.1038/s41586-020-2798-3

 

Hafner T., Popp D., and Zhang J., 2020, COVID-19 vaccines and intellectual property: recommendations for equity and global access, Health Affairs, 39(9): 1608-1614.

https://doi.org/10.1377/hlthaff.2020.01229

 

Karikó K., Muramatsu H., and Weissman D., 2015, Increased immune activation of nucleoside-modified mRNA for vaccine delivery, Cell, 162(5): 931-943.

https://doi.org/10.1016/j.cell.2015.07.017

 

Kranz L.M., Diken M., Haas H., Kreiter S., Loquai C., Reuter K.C., Meng M., Fritz D., Vascotto F., Hefesha H., Grunwitz C., Vormehr M., Hüsemann Y., Selmi A., Kuhn A.N.,Buck J., Derhovanessian E., Rae R., Sebastian Attig S., Diekmann J., Jabulowsky R.A., Heesch S., Jessica Hassel J., Langguth P., Stephan Grabbe S., Christoph Huber C., Türeci Ö., and Sahin U., 2016, Systemic RNA delivery to dendritic cells exploits antiviral defense for cancer immunotherapy, Nature, 534(7607): 396-401.

https://doi.org/10.1038/nature18300

PMID: 27281205

 

Mevorach D., Anis E., Cedar N., Bromberg, M., Haas, E. J., Nadir, E., Olsha-Castell S., Arad D., Hasin T., Levi N., Asleh R., Amir O., Meir K., Cohen D., Dichtiar R., Novick D., Hershkovitz Y., Dagan R., Leitersdorf I., Ben-Ami R., Miskin I., Saliba W., Muhsen K., Levi Y., Green M.S., Keinan-Boker L., and Alroy-Preis S., 2021, Myocarditis after BNT162b2 mRNA vaccine against COVID-19 in Israel, The New England Journal of Medicine, 385(23): 2140-2149.

https://doi.org/10.1056/NEJMoa2109730

PMID: 34614328 PMCID: PMC8531987

 

Moon S., Armstrong J., Hutler B., and Upshur R., 2021, COVAX and global equity: what can we learn from the past?, Globalization and Health, 17(1): 1-6.

https://doi.org/10.1186/s12992-021-00712-2

 

Pardi N., Hogan M.J., Porter F.W., and Weissman D., 2018, mRNA vaccines-A new era in vaccinology, Nature Reviews Drug Discovery, 17(4): 261-279.

https://doi.org/10.1038/nrd.2017.243

PMID: 29326426 PMCID: PMC5906799

 

Phelan A.L., Eccleston-Turner M., Rourke M., Maleche A., and Wang C.G., 2021, Legal agreements: barriers and enablers to global equitable COVID-19 vaccine access, The Lancet, 398(10297): 358-364.

https://doi.org/10.1016/S0140-6736(21)00922-3

PMID: 32910908 PMCID: PMC7832682

 

Polack F.P., Thomas S.J., Kitchin N., Absalon J., Gurtman A., Lockhart S., Perez J.L., Marc G.P., Moreira E.D., Zerbini C., Bailey R., Swanson K.A., Roychoudhury S., Koury K., Li P., Kalina W.V., Cooper D., Frenck Jr R.W., Hammitt L.L., Türeci Ö., Nell H., Schaefer A., Ünal S., Tresnan D.B., Mather S., Dormitzer P.R., Şahin U., Jansen K.U., Gruber W.C., and C4591001 Clinical Trial Group, 2020, Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine, The New England Journal of Medicine, 383(27): 2603-2615.

https://doi.org/10.1056/NEJMoa2034577

PMID: 33301246 PMCID: PMC7745181

 

Rauch S., Jasny E., Schmidt K.E., and Petsch B., 2018, New vaccine technologies to combat outbreak situations, Frontiers in Immunology, 9: 1963.

https://doi.org/10.3389/fimmu.2018.01963

PMID: 30283434 PMCID: PMC6156540

 

Roldão A., Picanço-Castro V., and Lobe M., 2021, Advances in non-viral mRNA delivery systems for vaccine applications, Frontiers in Bioengineering and Biotechnology, 9: 660085.

https://doi.org/10.3389/fbioe.2021.660085

 

Sahin U., and Türeci Ö., 2017, Personalized vaccines for cancer immunotherapy, Science, 359(6382): 1355-1360.

https://doi.org/10.1126/science.aar7112

PMID: 29567706

 

Sahin U., Muik A., Derhovanessian E., Vogler I., Kranz L.M., Vormehr M., Baum A., Pascal K., Quandt J., Maurus D., Brachtendorf S., Verena Lörks V., Sikorski J., Hilker R., Becker D., Eller A.K, Grützner J., Boesler C., Rosenbaum C., Kühnle M.C., Luxemburger U., Kemmer-Brück A., Langer D., Bexon M., Bolte S., Karikó K., Tania Palanche T., Fischer B., Schultz A., Shi P.Y., Fontes-Garfias C., Perez J.L., Swanson K.A., Loschko J.L., Scully I.L., Cutler M., Kalina W., Kyratsous C.A., Cooper D., Dormitzer P.R., Jansen K.U., and Türeci Ö., 2020, COVID-19 vaccine BNT162b1 elicits human antibody and TH1 T cell responses, Nature, 586(7830): 594-599.

https://doi.org/10.1038/s41586-020-2814-7

PMID: 32998157

 

Shin M.D., Shukla, S., Chung Y.H., Beiss V., Chan S.K., Ortega-Rivera O.A., and Steinmetz N.F., 2021, COVID-19 vaccine development and a potential nanomaterial path forward, Nature Nanotechnology, 15(8): 646-655.

https://doi.org/10.1038/s41565-020-0737-y

PMID: 32669664

 

Siewert C.D., Haas H., and Cornelis J.J., 2020, PLGA-based nanoparticles for mRNA delivery: past, present, and future, Journal of Controlled Release, 328: 759-774.

https://doi.org/10.1016/j.jconrel.2020.09.046

 

Tanne J.H., 2020, Covid-19: FDA approves Pfizer-BioNTech vaccine within days of application, BMJ, 371: m4707.

https://doi.org/10.1136/bmj.m4707

 

Thompson M.G., Burgess J.L., Naleway A.L., Tyner H., Yoon S.K., Meece J., Olsho L.E.W., Caban-Martinez A.J., Fowlkes A.L., Lutrick K., Groom H.C., Dunnigan K., Odean M.J., Hegmann K., Stefanski E., Edwards L.J., Schaefer-Solle N., Grant L., Ellingson K., Kuntz J.L, Zunie T., Thiese M.S., Ivacic L., Wesley M.G., Lamberte J.M., Sun X.X, Smith M.E., Phillips A.L., Groover K.D., Yoo Y.M., Gerald J., Brown R.T., Herring M.K., Joseph G., Beitel S., Morrill T.C., Mak J., Rivers P., Poe B.P., Lynch B., Zhou Y.T., Zhang J., Kelleher A., Li Y., Dickerson M., Hanson E., Guenther K., Tong S.X., Bateman A., Reisdorf E., Barnes J., Azziz-Baumgartner E., Hunt D.R., Arvay M.L., Kutty P., Fry A.M., and Gaglani M., 2021, Prevention and attenuation of COVID-19 with the BNT162b2 and mRNA-1273 vaccines, The New England Journal of Medicine, 385(4): 320-329.

https://doi.org/10.1056/NEJMoa2107058

 

Walsh E.E., Frenck R.W., Falsey A.R., Kitchin N., Absalon J., Gurtman A., Lockhart S., Neuzil K., Mulligan M.J., Bailey R., Swanson K.A., Ping Li P., Koury K., Kalina W., Cooper D., Fontes-Garfias C., Shi P.Y., Türeci Ö., Tompkins K.R., Lyke K.E., Raabe V., Dormitzer P.R., Jansen K.U., Şahin U., and Gruber W.C., 2020, Safety and immunogenicity of two RNA-based COVID-19 vaccine candidates, The New England Journal of Medicine, 383(25): 2439-2450.

https://doi.org/10.1056/NEJMoa2027906

PMID: 33053279 PMCID: PMC7583697

 

Wang W., 2024, Studying the dynamic changes of the immune system through single-cell omics, International Journal of Molecular Medical Science, 14(1): 29-41.

https://doi.org/10.5376/ijmms.2024.14.0005

 

Wouters O.J., Shadlen K.C., Salcher-Konrad M., Pollard A.J., Larson H.J., Teerawattananon Y., and Jit M., 2021, Challenges in ensuring global access to COVID-19 vaccines: production, affordability, allocation, and deployment, The Lancet, 397(10278): 1023-1034.

https://doi.org/10.1016/S0140-6736(21)00306-8

PMID: 33587887 PMCID: PMC7906643

Journal of Vaccine Research
• Volume 14
View Options
. PDF
. HTML
Associated material
. Readers' comments
Other articles by authors
. Xiaojie Zhang
Related articles
. mRNA vaccines
. Lipid nanoparticles
. COVID-19
. Cancer immunotherapy
. Global health
Tools
. Post a comment