Vaccination remains a cornerstone in preventing infectious diseases and managing outbreaks. The COVID-19 pandemic has underscored the revolutionary impact of mRNA vaccine technology, which utilizes pathogenderived genomic sequences to generate specific antigens. This process involves in vitro transcription of mRNA, encoding target antigens that are subsequently encapsulated within lipid nanoparticles (LNPs) for efficient delivery into host cells. Once internalized, the mRNA enables antigen expression, triggering a robust immune response. This platform dramatically accelerates vaccine development timelines and offers unparalleled adaptability, making mRNA vaccines particularly advantageous in addressing emerging infectious diseases. The clinical success of BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) has fueled broader applications, including influenza, respiratory syncytial virus (RSV), Zika, and HIV. Notably, mRNA-1345 became the first FDA-approved RSV mRNA vaccine, while self-amplifying RNA and multivalent vaccines are advancing in trials. However, CureVac’s CVnCoV failed due to lack of nucleoside modifications, and mRNA-1325 (Zika) showed poor immunogenicity. Additionally, mRNA-1365 (RSV) faced an FDA clinical hold due to safety concerns. These cases highlight the need for continued optimization in sequence design, delivery, and safety assessment. Despite advancements, a key hurdle persists, including mRNA instability, ultra-low storage requirements, and LNP liver accumulation. Innovations such as lyophilization and selective organ targeting technology are being explored to improve stability extrahepatic delivery. This review examines mRNA vaccine optimization strategies, clinical progress, and challenges, providing insights into future developments in this evolving field.

Vaccination remains one of humanity’s most powerful tools in the fight against infectious diseases. According to the World Health Organization, vaccines prevent an estimated million of deaths annually [1]. However, traditional vaccine development encounters several challenges, including lengthy production timelines and the rapid mutation of viruses, particularly RNA viruses, which can render vaccines ineffective against emerging strains [2]. Additionally, safety concerns such as the risk of ‘virulence reversion’, where live attenuated vaccines might mutate back to a pathogenic form, further complicate conventional vaccine development (Table 1) [3].

Table 1:
Comparison of prophylactic vaccine types for infectious diseases.
Vaccine typeImmune responseStabilitySafetyExamples
Live attenuated vaccines Robust humoral and cellular immunity Requires cold chain; moderate stability Risk of reversion to virulence; not for immunocompromised individuals Yellow fever,
oral polio 
Inactivated vaccines Primarily humoral immunity Stable; requires cold chain No reversion risk; lower immunogenicity Hepatitis A;
rabies 
Subunit vaccines Primarily humoral immunity High stable; no cold chain needed Very safe; requires adjuvants Hepatitis B;
HPV;
pertussis 
Viral vector vaccines Robust humoral and cellular immunity Requires cold chain; moderate stability Pre-existing immunity may reduce efficacy COVID-19 
LNP-mRNA vaccines Robust humoral and cellular immunity Requires ultra-low temperature storage; improving with lyophilization Possible reactogenicity;
concerns over LNP and PEG sensitivity 
COVID-19 
Vaccine typeImmune responseStabilitySafetyExamples
Live attenuated vaccines Robust humoral and cellular immunity Requires cold chain; moderate stability Risk of reversion to virulence; not for immunocompromised individuals Yellow fever,
oral polio 
Inactivated vaccines Primarily humoral immunity Stable; requires cold chain No reversion risk; lower immunogenicity Hepatitis A;
rabies 
Subunit vaccines Primarily humoral immunity High stable; no cold chain needed Very safe; requires adjuvants Hepatitis B;
HPV;
pertussis 
Viral vector vaccines Robust humoral and cellular immunity Requires cold chain; moderate stability Pre-existing immunity may reduce efficacy COVID-19 
LNP-mRNA vaccines Robust humoral and cellular immunity Requires ultra-low temperature storage; improving with lyophilization Possible reactogenicity;
concerns over LNP and PEG sensitivity 
COVID-19 

In recent decades, mRNA technology has emerged as a ground-breaking platform for vaccine and therapeutic development. Since the discovery of mRNA in 1961, its potential has been progressively explored, with early demonstrations of in vitro transcription (IVT) and successful validation in mice occurring in 1990s [4,5]. However, the clinical application of mRNA faced substantial hurdles, including poor cellular uptake due to its negatively charged phosphate backbone, susceptibility to enzymatic degradation, and rapid systemic clearance [6,7]. These challenges underscored the crucial need for an efficient and safe delivery system to fully unlock mRNA’s therapeutic potential.

The development of lipid-based delivery systems has been a major milestone in overcoming these obstacles. Liposomes were first explored for nucleic acid delivery in the 1970s, but their short half-life, high sensitivity, and a complex preparation process limited their practical use [8-10]. A significant breakthrough came in the 2010s with the development of four-component lipid nanoparticles (LNPs), which revolutionized nucleic acid delivery [11]. By 2018, the FDA had approved the first LNP-based siRNA therapeutic, paving the way for further advancements. The emergence of the SARS-CoV-2 pandemic in 2020 accelerated research into LNP-mRNA vaccines [12]. Compared with traditional vaccines, mRNA vaccines offer substantial advantages, such as bypassing the need for cell culture and enabling rapid large-scale production, dramatically reducing development timelines.

Despite these advantages, several challenges remain. Issues such as ultra-low temperature storage requirements, potential allergic reactions to LNP components, and the rapid waning of neutralizing antibody titers are the highlight areas that need further improvement [13-16]. Additionally, expanding mRNA vaccine distribution to extrahepatic tissues is another key frontier.

This review aims to provide a comprehensive analysis of the key components of LNP-mRNA vaccines, including antigen-encoding RNA design, LNP composition, and strategies for optimization. Furthermore, it will explore the current landscape of clinical trials for LNP-mRNA vaccines targeting infectious diseases, lessons from failed trails, and advances in extrahepatic tissues delivery, contributing to the ongoing advancements in this rapidly evolving field.

LNP-mRNA vaccines possess the remarkable ability to activate both the innate and the adaptive immune systems. Upon administration, exogenous mRNA that successfully enters cells is rapidly recognized by various pattern recognition receptors (PRRs), triggering a cascade of innate immune responses. Among these PRRs, Toll-like receptors 7 and 8 (TLR7/8) play a crucial role in detecting single-stranded RNA [17]. Additionally, double-stranded mRNA fragments can be sensed by TLR3 in the endosome, as well as cytoplasmic receptors such as retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). Activation of these diverse sensory pathways converges on shared downstream signaling nodes, culminating in the coordinated biosynthesis of type I interferons (primarily IFN-α/β) and pro-inflammatory cytokines. This inflammatory milieu functions as an ‘adjuvant-like’ environment, priming the immune system for subsequent adaptive responses.

Simultaneously, mRNA vaccines are internalized by local antigen-presenting cells (APCs), where they are translated into antigenic proteins. These proteins are then degraded into smaller peptides by the proteasome and presented on major histocompatibility complex class I (MHC-I) molecules to activate CD8+ cytotoxic T lymphocytes. In parallel, the antigenic peptides that enter the endosomal/lysosomal pathway are loaded onto MHC-II molecules for presentation to CD4+ helper T cells [18,19]. The expression of cytokines, such as type I interferons and co-stimulatory molecules, is crucial for APC-mediated T cell activation. Moreover, secreted antigenic proteins can bind to B cell receptors, triggering humoral immunity. CD4+ follicular helper T (Tfh) cells further assist B cells in producing high-affinity antibodies [20]. Research in animal models has demonstrated that LNPs induce high levels of IL-6 and other cytokines, which promote Tfh cells differentiation and germinal B cells formation, thereby accelerating the affinity maturation of antibodies. Moreover, type I interferon signaling enhances CD8+ T cells activation and proliferation, further strengthening immune protection [17,21].

In conclusion, mRNA vaccines effectively drive APCs maturation and lymphocytes activation. The innate immune system establishes an inflammatory environment rich in cytokines and interferons, providing a foundation for robust B and T cell responses. The coordinated activation of innate and adaptive immunity ultimately confers strong and durable protective immunity (Figure 1).

Immune responses induced by LNP-mRNA vaccines.

Figure 1:
Immune responses induced by LNP-mRNA vaccines.

Upon administration, LNPs facilitate mRNA uptake by antigen-presenting cells (APCs), leading to intracellular antigen expression. The mRNA or its byproducts can be recognized by pattern recognition receptors (PRRs) such as Toll-like receptors 7 and 8 (TLR7/8), retinoic acid-inducible gene I (RIG-I), and melanoma differentiation-associated protein 5 (MDA5), triggering an innate immune response characterized by the release of interferons (IFN-α/β) and pro-inflammatory cytokines. These signals enhance APC maturation, leading to the activation of CD4+ helper T cells (via MHC II presentation) and CD8+ cytotoxic T cells (via MHC I presentation), which collectively contribute to adaptive immune responses. Additionally, secreted antigenic proteins can activate B cells, leading to antibody production. This figure was created using BioRender.com.

Figure 1:
Immune responses induced by LNP-mRNA vaccines.

Upon administration, LNPs facilitate mRNA uptake by antigen-presenting cells (APCs), leading to intracellular antigen expression. The mRNA or its byproducts can be recognized by pattern recognition receptors (PRRs) such as Toll-like receptors 7 and 8 (TLR7/8), retinoic acid-inducible gene I (RIG-I), and melanoma differentiation-associated protein 5 (MDA5), triggering an innate immune response characterized by the release of interferons (IFN-α/β) and pro-inflammatory cytokines. These signals enhance APC maturation, leading to the activation of CD4+ helper T cells (via MHC II presentation) and CD8+ cytotoxic T cells (via MHC I presentation), which collectively contribute to adaptive immune responses. Additionally, secreted antigenic proteins can activate B cells, leading to antibody production. This figure was created using BioRender.com.

Close modal

LNP-mRNA vaccines can be categorized based on RNA type. Currently, three major forms of RNA can be produced via IVT: conventional linear mRNA, self-amplifying RNA (saRNA), and circular RNA (circRNA) (Figure 2) [22].

Types of RNA synthesized through in vitro transcription.

Figure 2:
Types of RNA synthesized through in vitro transcription.
Figure 2:
Types of RNA synthesized through in vitro transcription.
Close modal

Conventional mRNA, the most widely used type, consists of a 5´ cap, a 5´ untranslated region (UTR), an open reading frame (ORF) encoding the antigen, a 3´ UTR, and a poly (A) tail. While IVT technology enables rapid intracellular protein expression, conventional mRNA is highly susceptible to enzymatic degradation and requires high doses to achieve protective efficacy.

saRNA, derived from positive-sense RNA viruses, represents the next generation of RNA technology. By encoding a replicase while replacing viral structural genes with antigen sequences, saRNA can self-replicate within cells, significantly reducing the required vaccine dose and administration frequency [23]. This mitigates challenges such as the ‘PEG dilemma’ observed in repeated vaccinations [24]. Researchers have indicated that saRNA vaccines elicit robust immune responses at much lower doses than conventional mRNA vaccines. For instance, McKay et al. developed an saRNA-based SARS-CoV-2 vaccine that induced strong antibody titers, a Th1-biased immune response, and superior neutralization against pseudoviruses and wildtype strains, exceeding the antibody potency found in convalescent COVID-19 patients [25].

Additionally, saRNA generates immunostimulatory signals via dsRNA intermediates formed during replication, further enhancing the innate immune response [26-28]. Nevertheless, saRNA molecules are relatively large, typically exceeding 9 kb as they encompass both replicase and antigen sequences. This characteristic imposes higher performance requirements on delivery vectors. Their production process is more complex than conventional mRNA. Moreover, while saRNA-induced innate immunity can enhance immunogenicity, excessive activation may suppress antigen expression, requiring careful optimization [27].

circRNA, characterized by a closed-loop structure lacking a 5´ cap and 3´ poly (A) tail, exhibits enhanced nuclease resistance and prolonged in vivo stability [29]. Notably, circRNA vaccines remain at 4℃ for at least four weeks, whereas conventional linear mRNA-LNP vaccines degrade under similar conditions within three weeks [30]. Due to their closed structure, circRNAs evade innate immune recognition more effectively, reducing excessive inflammatory responses and potentially improving safety [31]. Besides, studies have demonstrated that engineered and optimized circRNA exhibits remarkable superiority in protein production. In vitro, the level of active protein generated by circRNA is substantially higher than that produced by an equivalent amount of linear mRNA. This advantage is observed not only in vitro but also in vivo, where circRNA shows both enhanced translational efficiency and prolonged protein expression [32].

Advancements in delivery systems have further expanded the potential of circRNA vaccines. For instance, a mannose-modified LNP vector has been developed to enhance lymph node targeting, leading to prolonged antigen availability and stronger Tfh and germinal center B cell responses. Mouse studies revealed that lyophilized mLNP-circRNA vaccines retained stability at 4℃ for up to 24 weeks without loss of immunogenicity [33]. These findings suggest that by enhancing delivery methods, such as targeted delivery and improved stability during lyophilization, circRNA vaccines can offer greater convenience for storage and transportation while preserving high efficacy. The concept of circRNA vaccines has been validated in multiple disease models. For example, Wei et al. developed a circRNA vaccine encoding the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein. In studies with macaque monkey experiments, this vaccine successfully elicited high-titer neutralizing antibodies and a significant T cell response, and it offered effective protection against both Delta- and Omicron-mutant strains [30]. Despite these promising advances, circRNA vaccines remain in the early stages of research, with key challenges such as large-scale production, in vivo metabolism, and long-term safety requiring further investigation [34].

Enhancing the stability, reducing immunogenicity, and improving protein expression of mRNA can be achieved through various optimization strategies. Key approaches include nucleoside modification, codon optimization, and structural refinement [35-37].

Nucleoside modification

A major challenge of exogenous mRNA is its innate immunogenicity, which can trigger excessive inflammatory responses and interfere with antigen expression, potentially reducing vaccine efficacy and causing adverse effects [38]. Researchers have indicated that inappropriate or excessive activation of type I interferons, such as IFN-α, may disrupt antigen expression, reduce vaccine effectiveness, and potentially lead to severe adverse effects [39]. To mitigate this, various chemical modifications have been introduced to reduce immunogenicity while maintaining translation efficiency [40]. For instance, natural adenosine can be replaced with N1-methyladenosine (m1A) or N6-methyladenosine (m6A), and cytosine can be substituted with 5-methylcytosine (5-mC) [41,42]. Additionally, uridine can be replaced with 5-methoxyuridine (5moU), pseudouridine (Ψ), or N1-methylpseudouridine (m1Ψ) [43]. Notably, both 5-mC and Ψ have been shown to reduce mRNA immunogenicity and enhance protein translation in both in vivo and in vitro studies [44].

Optimization of codon

Codon usage can be tailored to host cell preferences to enhance translation efficiency and protein yield. Strategies include substituting rare codons with more prevalent ones (to prevent ribosomal stalling) and increasing GC content to improve stability. For instance, codon-optimized E7 genes have shown enhanced immunotherapeutic efficacy in HPV-16 vaccines [45]. COVID-19 . After purifiedmRNA vaccines developed by Moderna and Pfizer/BioNTech, codon optimization improved antigen expression and vaccine effectiveness [46]. Additionally, optimizing codon sequences can reduce unwanted activation by minimizing RIG-I-mediated innate immune activation [47].

Optimization of non-coding regions

The non-coding regions of mRNA, including the 5´ cap, 5´ UTR, 3´ UTR, and 3´ Poly (A) tail, play essential roles in regulating mRNA stability, translation, and cellular function.

5´ cap particularly the m7G cap structure (cap-0, cap-1, and cap-2) protects mRNA from exonuclease degradation and enhances translation efficiency while reducing immunogenicity [48].

UTRs influence translation efficiency, stability and localization. Using highly expressed natural UTRs, such as α- and β-globin, can prevent degradation and improve mRNA longevity [49].

Poly (A) tail, its length typically around 100 nt, is crucial for mRNA stability and expression efficiency. However, studies suggest that a 75 nt poly (A) tail may achieve optimal expression in human cells, indicating a context-dependent balance [50,51]. Novel designs, such as segmented poly (A) tail, further extend stability and translation efficiency [52].

Artificial intelligence-assisted mRNA optimization

Machine learning and deep learning algorithms are revolutionizing mRNA vaccine design by enabling precise sequence optimization.

Advanced models such as convolutional neural networks, residual neural networks, and graph neural networks can analyze large-scale sequence data to extract intricate features, including codon usage preferences and secondary structures. Leveraging these deep learning architectures, researchers can subsequently optimize mRNA stability and translation efficiency by fine-tuning its design based on the complex patterns identified [53]. For example, during COVID-19 vaccine development, researchers introduced the LinearDesign algorithm, which optimized mRNA stability and codon usage within 11 minutes, leading to a 128-fold increase in antibody titers in mice [54]. Additionally, machine learning models have been employed to fine-tune UTR design and stabilize ORF structures (CDSFold model) to enhance vaccine efficacy [55]. Epitope prediction is a cornerstone of mRNA vaccine design. As the specific regions on antigen proteins where antibodies bind, epitopes play a critical role in vaccine efficacy, making their precise identification essential. Commonly used prediction tools, such as DeepAb, BepiPred, and DiscoTope, employ deep learning algorithms to analyze complex datasets [56]. By integrating computational techniques with bioinformatics, these tools efficiently screen for candidate sequences with robust immunogenic potential, thereby significantly enhancing both the precision and efficiency of mRNA vaccine development.

LNPs are the most widely used delivery systems for mRNA vaccines, demonstrating efficient mRNA protection and intracellular delivery during the COVID-19 pandemic (Figure 3). Typically composed of ionizable lipids, cholesterol, helper lipids, and PEGylated lipids, LNPs enable rapid, scalable production with high biosafety. However, challenges such as hepatic accumulation, potential immunogenicity, and targeted delivery limitations remain. Addressing these issues is crucial for expanding LNP applications in RNA vaccines.

Preparation of LNP-mRNA vaccines.

Figure 3:
Preparation of LNP-mRNA vaccines.

The DNA transcription template is designed based on the target antigen sequence, and then undergoes in vitro transcription (IVT) to generate mRNA. After purified, mRNA is subsequently mixed with lipid components to form LNP-mRNA complexes. Following LNP encapsulation, the formulation undergoes buffer exchange and sterile filtration to ensure safety and purity before being filled into vials for clinical use and distribution. This figure was created using BioRender.com.

Figure 3:
Preparation of LNP-mRNA vaccines.

The DNA transcription template is designed based on the target antigen sequence, and then undergoes in vitro transcription (IVT) to generate mRNA. After purified, mRNA is subsequently mixed with lipid components to form LNP-mRNA complexes. Following LNP encapsulation, the formulation undergoes buffer exchange and sterile filtration to ensure safety and purity before being filled into vials for clinical use and distribution. This figure was created using BioRender.com.

Close modal

Ionizable lipids

Ionizable lipids (ILs), constituting 30–50% of LNP composition, play a pivotal role in RNA encapsulation and endosomal escape. They typically contain a hydrophilic ionizable cationic headgroup, a biodegradable linker, and one or more hydrophobic tails [57]. Their pKa (typically 6–7) is optimized to facilitate RNA binding in acidic environments while remaining neutral at physiological pH, minimizing toxicity [58]. When LNPs are internalized by cells via endocytosis, the acidic environment of the endosomes encourages fusion with the endosomal membrane, ensuring efficient release of the mRNA cargos into the cytoplasm.

DODAP and DODMA are among the first ILs used for RNA delivery [59]. The first FDA-approved siRNA-LNP therapeutic, patisiran (Onpattro®), utilized Dlin-MC3-DMA ILs [60]. This breakthrough led to further IL innovations, including ALC-0315 (Pfizer/BioNTech BNT162b2) and SM-102 (Moderna mRNA-1273) [61]. ILs such as Dlin-MC3-DMA and L-608 (Merck 32) have been evaluated in respiratory syncytial virus (RSV) vaccine trials [62]. And ATX-100 was used in Arcturus' COVID-19 saRNA vaccine ARCT-154 [63]. In addition, some ILs remain undisclosed, such as those in the ARCoV vaccine co-developed by Suzhou Abogen, Walvax and the Academy of Military Medical Sciences of China (Figure 4) [64].

LNP components used in vaccines and vaccine candidates.

Figure 4:
LNP components used in vaccines and vaccine candidates.
Figure 4:
LNP components used in vaccines and vaccine candidates.
Close modal

Recent IL advancements include polycyclic adamantane-based ILs (e.g. 11-AM) and cyclic imidazole-modified ILs (e.g. 93-O17S), which show potential for T cell targeting [65]. Furthermore, A18-Iso5-2DC18, with a cyclic amine headgroup, exhibits strong STING activation, promoting dendritic cell maturation [66]. Notably, an LNP incorporating 4N4T (four tertiary amine nitrogen atoms and four hydrophobic tails) lipids elicited stronger Th1-based immune responses and higher RBD-specific IgG and neutralizing antibody titers than SM-102-based vaccines [67].

Cholesterol

Cholesterol, comprising 20–50% of LNPs, enhances membrane stability, delivery efficiency, and circulatory half-life [68]. Natural cholesterol is commonly used in current mRNA vaccines, but alternatives like β-sitosterol, a natural analogue, may further boost transfection efficiency by influencing cellular uptake [69]. However, the long-term safety of β-sitosterol and the costs associated with large-scale production still require further evaluation.

Helper lipids

Helper lipids, the most commonly used are phospholipids, typically 10–20% of LNPs, forming a bilayer membrane that stabilizes RNA encapsulation and promotes efficient delivery. Saturated phosphatidylcholine lipids, such as 1,2-distearoyl-sn-glycero-3-phosphocholine, are widely used in RNA vaccines. In addition to their critical role in membrane formation, altering the polarity of phospholipid has been shown to influence the organ-targeting characteristics of LNPs [70].

PEGylated lipids

PEGylated lipids make up a small fraction of the LNPs, typically around 1.5%. Despite their low abundance, increasing the content of PEGylated lipids improves the size distribution and overall stability of LNPs. This enhancement provides a protective shield against protein adsorption in the bloodstream, which in turn prevents recognition and clearance by the reticuloendothelial system and the mononuclear phagocyte system, thereby extending the circulation half-life of LNPs [68,71]. Research indicates that the length of the carbon chain in these PEGylated lipids affects their desorption rate. Presently, commercially available RNA vaccines use PEGylated lipids with 14-carbon chains, such as the ALC-0159 and DMG-PEG2000.

However, the widespread use of PEG in consumer products has led to pre-existing anti-PEG antibodies (APAs) in the population, which may accelerate LNP clearance (accelerated blood clearance, ABC), potentially reducing vaccine efficacy [72]. Repeated administration might further compromise vaccine effectiveness. Notably, between December 14 and 23, 2020, clinical data reported in the Vaccine Adverse Event Reporting System identified 21 cases of anaphylaxis in 1.89 million Pfizer/BioNTech vaccines. These reactions are most likely associated with sensitivity to PEG [73].

To address the challenges associated with PEG dilemma, various strategies have been explored, including enhancing mRNA expression efficiency to reduce dose requirements, replacing mRNA with saRNA that requires lower doses, and substituting PEGylated lipids with alternative materials. For example, Dong et al. demonstrated that replacing PEGylated lipids with pSar lipids improved mRNA delivery while maintaining safety [61]. Optimizing PEG terminal groups has also emerged as a promising strategy. Methoxy-PEGylation (MeO-PEG) is widely applied in numerous marketed nanomedicines, including MeO-PEG-DMG and ALC-0159 [74,75]. Zhan et al. discovered that hydroxyl-PEGylation (OH-PEG) at the PEG terminus effectively prevents binding by pre-existing APAs in humans [76]. Another innovative approach is substituting PEG with brush-shaped polymer lipids (BPLs), which exhibit lower APA affinity, prolonged circulation, and enhanced mRNA delivery. Xiao et al. developed high-density BPLs, demonstrating superior performance over conventional DMG-PEG2000 LNPs, particularly in repeated-dose studies [77]. These advancements provide new strategies to mitigate PEG-related limitations, paving the way for safer and more effective LNP-mRNA vaccines.

The recent achievements of mRNA vaccines, as seen in bnt162b2 (Pfizer-Biontech) and mRNA-1273 (Moderna), have sparked interest in their application to various infectious diseases like influenza, RSV, Zika virus, HIV, and more (Table 2). With their swift design, scalable production, and targeted immunogenicity, mRNA vaccines hold promise in addressing persistent challenges in vaccine development, presenting effective solutions for both emerging and re-emerging infectious diseases.

Table 2:
Global clinical trials of RNA-based prophylactic vaccines for infectious diseases.
PathogenClinicalTrials.Gov IDLatest R&D statusVaccine nameSponsorMain purpose
COVID-19 NCT05375838 Phase I/II Elasomeran, mRNA-1073,mRNA-1010 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1073 (SARS-CoV-2 and Influenza Vaccine). Compared with co-administered mRNA-1010 (Influenza) in healthy adults 18–75 years of age 
 NCT06354998 Phase III Elasomeran, mRNA-1273.815 ModernaTX, Inc. Randomized, observer-blind, active-controlled of mRNA-1273.815 COVID-19 vaccine in previously vaccinated adults 
 NCT05137236 Phase II Elasomeran, mRNA-1283, mRNA-1283.211, mRNA-1283.529 ModernaTX, Inc. Immunogenicity and safety of mRNA-1283 vaccine boosters for SARS-CoV-2 
 NCT04927065 Phase II/III Elasomeran, elasomeran + imelasomeran, mRNA-1273.213, mRNA-1273.815, mRNA-1273.231, mRNA-1273.211, mRNA-1273.529, elasomeran + davesomeran, mRNA-1273.617.2 ModernaTX, Inc. Immunogenicity and safety of mRNA vaccine boosters for SARS-CoV-2 variants 
 NCT04796896 Phase II/III Elasomeran, elasomeran, imelasomeran ModernaTX, Inc. Safety, tolerability, reactogenicity, and effectiveness of mRNA-1273 SARS-CoV-2 vaccine in healthy children 6 months to less than 12 years of age 
 NCT04649151 Phase II/III Elasomeran, elasomeran + davesomeran ModernaTX, Inc. Safety, reactogenicity, and effectiveness of mRNA-1273 SARS-CoV-2 vaccine in healthy adolescents 12 to < 18 years of age 
 NCT04380701 Phase I/II BNT162a1, abdavomeran, tozinameran, pidacmeran BioNTech SE Safety and immunogenicity of four prophylactic SARS-CoV-2 RNA vaccines against COVID-19 using different dosing regimens in healthy and immunocompromised adults 
 NCT05004181 Phase II BNT162b2 (B.1.617.2), BNT162b2 (B.1.1.7), BNT162b2 (B.1.1.7 + B.1.617.2) BioNTech SE A phase II trial to evaluate the safety and immunogenicity of SARS-CoV-2 monovalent and multivalent RNA-based vaccines in healthy subjects 
 NCT04649021 Phase II Tozinameran BioNTech SE Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (BNT162b2) in Chinese healthy population: A phase ii, randomized, placebo-controlled, observer-blinded study 
 NCT04537949 Phase I/II Ganulameran BioNTech SE Safety and immunogenicity of a prophylactic SARS-CoV-2 RNA vaccine (BNT162b3) against COVID-19 using different dosing regimens in healthy adults 
 NCT04523571 Phase I Abdavomeran BioNTech SE Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (BNT162b1) in Chinese healthy subjects: A phase i, randomized, placebo-controlled, observer-blind study 
 NCT04368728 Phase II/III BNT162b2s01, abdavomeran, tozinameran BioNTech SE, Pfizer Safety, tolerability, immunogenicity, and efficacy of SARS-CoV-2 RNA vaccine candidates against COVID-19 in healthy individuals 
 NCT04955626 Phase III Riltozinameran, tozinameran + riltozinameran, tozinameran BioNTech SE, Pfizer Additional dose(s) of BNT162b2 in healthy individuals previously vaccinated with BNT162b2 
 NCT06178991 Phase III PF-07926307 BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of a combined modified RNA vaccine candidate against COVID-19 and influenza in healthy individuals 
 NCT05472038 Phase II/III Tozinameran + riltozinameran, tozinameran + famtozinameran,BNT162b5, tozinameran BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of BNT162b RNA-based vaccine candidates in COVID-19 vaccine-experienced healthy individuals 
 NCT06279871 Phase III ARCT-2303 Arcturus Therapeutics, Inc. Immunogenicity, reactogenicity, and safety of a saRNA COVID-19 vaccine (ARCT-2303), administered concomitantly with quadrivalent influenza vaccines, in adults 
 NCT05037097 Phase I/II ARCT-021, ARCT-165, ARCT-154 Arcturus Therapeutics, Inc. Safety, reactogenicity, and immunogenicity of 3 SARS-CoV-2 RNA vaccine candidates in adults previously vaccinated and not previously vaccinated against SARS-CoV-2 
 NCT05012943 Phase II/III ARCT-154 Vinbiocare Biotechnology Joint Stock Company Immunogenicity and efficacy of the SARS-CoV-2 saRNA vaccine ARCT-154 in adults 
 NCT05517642 Phase III Ad5-nCoV-IH, BNT162b2 CanSino Biologics Inc. Immunogenicity, efficacy and safety of inhaled (IH) viral vectored vaccine (Convidecia, CanSino) as second booster dose against emerging variants of concern (VOC) of SARS-CoV-2 to prevent breakthrough infections 
 NCT05373472 Phase II CS-2034 CanSino Biologics Inc. Safety and immunogenicity of COVID-19 mRNA vaccine in adults aged 18 years and older 
 NCT05743335 Phase I JCXH-221 Immorna Biotherapeutics, Inc. Safety and immunogenicity of JCXH-221, an MRNA-based broadly protective COVID-19 vaccine 
 NCT04863131 Phase I/II EXG-5003 Fujita Health University Safety and immunogenicity of intradermal SARS-CoV-2 vaccine EXG-5003 in healthy adults 
 NCT05960097 Phase II CV0701, CV0601 GlaxoSmithKline, CureVac Safety, reactogenicity, and immunogenicity of a booster dose of investigational COVID-19 mRNA vaccines in healthy adults who previously received a complete primary vaccination series with or without booster dose(s) 
 NCT05477186 Phase I CV0501 GlaxoSmithKline Safety and immunogenicity study of a booster dose of the investigational CV0501 mRNA COVID-19 vaccine in adults at least 18 years old 
 NCT04758962 Phase I GSK4184258A GlaxoSmithKline Safety, reactogenicity and immune response of a CoV-2 SAM (LNP) vaccine when administered intramuscularly on a 0, 1 month schedule in healthy adults 18 to 50 years of age 
 NCT04652102 Phase II/III CVnCoV CureVac Efficacy and safety of investigational SARS-CoV-2 mRNA vaccine CVnCoV in adults 18 years of age and older 
 NCT05903118 Phase I/II RBMRNA-176 Argorna Pharmaceuticals Co., LTD Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (RBMRNA-176) in healthy adults aged 18 years and older 
 NCT05897190 Phase I RBMRNA-405, CoronaVac® Argorna Pharmaceuticals Co., LTD Safety and immunogenicity of a SARS-CoV-2 mRNA vaccine (RBMRNA-405) as a booster in Chinese adults and older 
 NCT05435027 Phase I GRT-R912, GRT-R914, GRT-R918 Gritstone bio, Inc. Safety and tolerability of GRT-R912, GRT-R914, and GRT-R918 administered as prime and/or boost in healthy adult participants and people living with HIV 
 NCT05148962 Phase I GRT-R910 Gritstone bio, Inc. Safety, immunogenicity, and reactogenicity of a self-amplifying mRNA prophylactic vaccine boost against SARS-CoV-2 in previously vaccinated healthy adults 18 years and older 
 NCT05329220 Phase III ABNCoV2, tozinameran Bavarian Nordic Immunogenicity, safety, and tolerability of a single dose of ABNCoV2 vaccine in adult subjects previously vaccinated for SARS-CoV-2: A phase 3 trial in two parts-randomized, double-blind, active controlled and open-label, single-arm 
 NCT05210179 Phase II Tozinameran, Turkovac Health Institutes of Turkey Safety, efficacy, and immunogenicity of booster vaccination (TURKOVAC) against SARS-CoV-2 
 NCT05668065 Phase III CoronaVac, CoronaVac, BNT162b2 Institut Pasteur de Tunis Immunogenicity of heterologous versus homologous prime boost schedule with mRNA and inactivated COVID-19 vaccines: A single-blinded, randomized, parallel group superiority trial 
 NCT05352867 Phase II LVRNA009 AIM Vaccine Co., Ltd. Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (LVRNA009) in Chinese people aged 18–59 years 
 NCT05343871 Phase IV Abdavomeran, ChAdOx1 nCoV-19, tozinameran Albert B. Sabin Vaccine Institut Immunogenicity and safety of full versus fractional dose of Pfizer/BioNTech, AstraZeneca, and Sinovac COVID-19 vaccines given as a booster dose at least 6 months after primary vaccination series or PCR-confirmed infection with SARS-CoV-2 in healthy adults 
 NCT05188677 Phase III SCB-2019, BNT162b2 Vaccine, Vaxzevria Vaccine, CoronaVac Vaccine Clover Biopharmaceuticals AUS Pty Ltd Immunogenicity and safety of adjuvanted recombinant SARS-CoV-2 trimeric S-protein subunit vaccine (SCB-2019), administered as a booster dose to adults, who previously received primary series of a COVID-19 vaccine 
 NCT04775069 Phase IV RBMRNA-405, CoronaVac® Humanity & Health Medical Group Limited Comparing the antibody response of subjects with chronic liver disease to mRNA, inactivated virus and adenovirus vector COVID-19 vaccines 
 NCT05030974 Phase IV Elasomeran, JNJ-78436735 University Medical Center Groningen Randomized multicenter study by the REnal patients COVID-19 VACcination (RECOVAC) Consortium 
 NCT04566276 Phase I/II ChulaCov19 Chulalongkorn University Safety, tolerability, and immunogenicity of the ChulaCov19 vaccine in healthy adults 
 NCT04569383 Phase I MVA-SARS-2-S, tozinameran Universitätsklinikum Hamburg-Eppendorf Safety, tolerability and immunogenicity of two ascending doses of the candidate vaccine MVA-SARS-2-S and heterologous booster vaccinations with a licensed vaccine against COVID-19 
 NCT04765436 Phase I PTX-COVID19-B Providence Therapeutics Holdings Inc. Safety, tolerability, and immunogenicity of PTX-COVID19-B vaccine in healthy seronegative adults aged 18–64 
 NCT04821674 Phase I/II DS-5670 Daiichi Sankyo Co., Ltd. Safety, immunogenicity and recommended dose of DS-5670a (COVID-19 vaccine) in Japanese healthy adults and elderly subjects 
 NCT05305573 Phase II Bimervax, tozinameran Hipra Scientific, S.L.U Immunogenicity and safety of a booster vaccination with a recombinant protein RBD fusion dimer candidate (PHH-1V) against SARS-CoV-2, in adults fully vaccinated with adenovirus vaccine against COVID-19 
 NCT05197153 Phase II Elasomeran, ChAdOx1 nCoV-19 Medigen Vaccine Biologics Corp. Safety, tolerability, and immunogenicity of heterologous booster dose with AZD1222, mRNA-1273, or MVC-COV1901 COVID-19 vaccine in adults 
 NCT05175742 Phase II PTX-COVID19-B, tozinameran Providence Therapeutics Holdings Inc. Safety, tolerability, and immunogenicity of PTX-COVID19-B compared to Pfizer-BioNTech COVID-19 vaccine in healthy seronegative adults aged 18 to 64 years 
 NCT05144139 Phase I/II SW0123 Stemirna Therapeutics Safety, immunogenicity and immune persistence of COVID-19 mRNA vaccine in healthy people aged 18 years and above 
 NCT05049226 Phase II ChAdOx1 nCoV-19, tozinameran Mahidol University Immunogenicity and safety of third dose vaccination with AstraZeneca COVID-19 vaccine or Pfizer/BioNTech COVID-19 vaccine among Thai Adults receiving two doses of Sinovac 
COVID-19, influenza nct06097273 Phase III mRNA-1083 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1083 (SARS-CoV-2 and influenza) vaccine in healthy adult participants, ≥50 years of age 
 NCT05596734 Phase I/II PF-07252220, PF-07926307, tozinameran + famtozinameran BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of combined modified RNA vaccine candidates against COVID-19 and influenza in healthy individuals 
 NCT04969276 Phase II Elasomeran, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of Fluzone® high-dose quadrivalent (influenza vaccine), 2021–2022 formulation and a third dose of Moderna COVID-19 vaccine (mRNA-1273 vaccine) administered either concomitantly or singly in adults 65 years of age and older previously vaccinated with a 2-dose schedule of Moderna COVID-19 vaccine 
 NCT05970887 Phase IV Tozinameran + famtozinameran Catholic Kwandong University Immunogenicity and safety of concomitant administration of Omicron-containing bivalent COVID-19 vaccines with seasonal influenza vaccines 
COVID-19, influenza, RSV NCT05585632 Phase I mRNA-1045, elasomeran, elasomeran + imelasomeran, mRNA-1010, mRNA-1230, mRNA-1345 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of multi-component vaccines mRNA-1045 (influenza and RSV) or mRNA-1230 (influenza, RSV, and SARS-CoV-2) compared with mRNA-1010 (influenza), mRNA-1345 (RSV), and mRNA-1273.214 (SARS-CoV-2) vaccines in healthy adults 50–75 years of age 
NCT05330975 Phase III Elasomeran, elasomeran + imelasomeran, Afluria Quadrivalent, Afluria (trivalent), mRNA-1345 ModernaTX, Inc. Safety, tolerability, and immunogenicity of mRNA-1345, an mRNA vaccine targeting respiratory syncytial virus (RSV), when given alone or coadministered with a seasonal influenza vaccine or SARS-CoV-2 vaccine and when given as an open-label boost at 1 year following a primary dose in adults ≥ 50 years of age 
Covid-19, RSV NCT05886777 Phase II PF-06928316, tozinameran + famtozinameran Pfizer Safety, tolerability, and immunogenicity of combined vaccine candidate(s) against infectious respiratory illnesses, including COVID-19 and RSV, in healthy individuals 
Covid-19, HPV NCT05119855 Phase III Elasomeran, Gardasil 9 Merck Sharp & Dohme LLC Safety and immunogenicity of 2-dose regimens of 9vhpv and mRNA-1273 SARS-CoV-2 vaccines where the first dose of each vaccine are given concomitantly in boys and girls 9 to 11 Years of age 
COVID-19, S. pneumoniae NCT05158140 Phase III mRNA-1273, V110, V114 Merck Sharp & Dohme LLC Safety, tolerability, and immunogenicity of the concomitant administration of either 23-valent pneumococcal polysaccharide vaccine or 15-valent pneumococcal conjugate vaccine with a booster dose of SARS-CoV-2 mRNA vaccine in healthy adults 50 years of age or older. 
 NCT04887948 Phase III 20vPnC Pfizer Safety and immunogenicity of 20 valent pneumococcal conjugate vaccine when coadministered with a booster dose of BNT162b2 in adults 65 Years of age and older 
Influenza NCT05540522 Phase III PF-07252220 Pfizer Efficacy, safety, tolerability, and immunogenicity of a modified RNA vaccine against influenza compared to licensed inactivated influenza vaccine in healthy adults 18 years of age or older 
 NCT05227001 Phase I PF-07836391, PF-07836395, PF-07836396, PF-07836394, PF-07852352, PF-07867246 Pfizer Safety, tolerability, and immunogenicity of saRNA vaccine preparations against influenza in healthy individuals 
 NCT03076385 Phase I VAL-506440 ModernaTX, Inc. Safety and immunogenicity of H10N8 antigen mRNA in healthy adult subjects 
 NCT05827978 Phase III mRNA-1010, Fluarix Quadrivalent (FLU D-QIV) ModernaTX, Inc. Immunogenicity, reactogenicity and safety of mRNA-1010 seasonal influenza vaccine in adults 18 years and older 
 NCT05333289 Phase I/II mRNA-1030, mRNA-1010, mRNA-1020 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1020 and mRNA-1030 candidate seasonal influenza vaccines in healthy adults 
 NCT03345043 Phase I VAL-339851 ModernaTX, Inc. Safety and immunogenicity of VAL-339851 in healthy subjects 
 NCT05566639 Phase III mRNA-1010 ModernaTX, Inc. Safety and efficacy of mRNA-1010 candidate seasonal influenza vaccine in adults 50 years and older 
Influenza NCT05566639 Phase III mRNA-1010 ModernaTX, Inc. Safety and efficacy of mRNA-1010 candidate seasonal influenza vaccine in adults 50 years and older 
 NCT05972174 Phase I/II mRNA-1018 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1018 pandemic influenza candidate vaccines in healthy adults ≥ 18 years of age 
 NCT05827068 Phase I/II mRNA-1012.1, mRNA-1010.3, mRNA-1011.2, mRNA-1010, mRNA-1011.1, mRNA-1010.2 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1011.1, mRNA-1011.2, and mRNA-1012.1 candidate seasonal influenza vaccines in healthy adults 50 to 75 years of age 
 NCT05868382 Phase II mRNA-1010.4, mRNA-1010, mRNA-1010.6 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mrna vaccine candidate variations in healthy adults 18 to 49 years of age 
 NCT06028347 Phase I SQ012 Seqirus Safety, reactogenicity and immunogenicity of an investigational self-amplifying MRNA influenza vaccine in healthy adults 
 NCT05829356 Phase I H3 mRNA/LNP Vaccine (sanofi), Flublok Quadrivalent Sanofi Pasteur, a Sanofi Company Early safety data reviews to assess safety and immunogenicity of one monovalent modified influenza mRNA vaccine encapsulated in LNP, in adults aged 18 to 49 years and 60 years and above. 
 NCT05650554 Phase I/II Fluzone Quadrivalent, MRT5413, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5413 in adults aged 18 Years and older 
 NCT05553301 Phase I/II MRT5407, Fluzone Quadrivalent, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5407 in adults aged 18 Years and older 
 NCT05624606 Phase I/II Fluzone Quadrivalent, MRT5410, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5410 in adults aged 18 years and older 
 NCT05426174 Phase I SP0273, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of monovalent mRNA vaccine in adult participants 18 years of age and older 
 NCT03814720 Phase I VRC-FLUNPF099-00-VP NIAID Dose, safety, tolerability, and immunogenicity of an influenza H1 stabilized stem ferritin vaccine, VRCFLUNPF099-00-VP, in healthy adults 
 NCT05446740 Phase I FLU SV mRNA GlaxoSmithKline, CureVac Safety, reactogenicity and immunogenicity of an mRNA-based monovalent influenza vaccine candidate in healthy younger and older adults 
 NCT05252338 Phase I CVSQIV GlaxoSmithKline, CureVac Safety, reactogenicity and immunogenicity of the investigational seasonal quadrivalent influenza mRNA vaccine CVSQIV administered intramuscularly in healthy younger and older adults 
Influenza, RSV NCT05788237 Phase I PF-07252220, PF-06928316 Pfizer Safety, tolerability, and immunogenicity of respiratory combination vaccine candidates in older adults 
NCT06060457 Phase III Fluzone High-Dose Quadrivalent, mRNA-1345 ModernaTX, Inc. Safety, tolerability, and immunogenicity of mRNA-1345, an mRNA vaccine targeting respiratory syncytial virus, when coadministered with a high-dose, quadrivalent seasonal influenza vaccine in adults ≥ 65 years of age 
HPIV, hMPV NCT04144348 Phase I mRNA-1653 ModernaTX, Inc. Safety and immunogenicity of mRNA-1653, a combined human metapneumovirus (hMPV) and parainfluenza virus type 3 (PIV3) vaccine when administered to adults, and to children 12 to 59 months of age with serologic evidence of prior exposure 
 NCT03392389 Phase I mRNA-1653 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1653, a combined human metapneumovirus and human parainfluenza virus type 3 vaccine, when administered to healthy adults 
ZIKV NCT04917861 Phase II mRNA-1893 ModernaTX, Inc. Safety, tolerability, and immunogenicity of Zika vaccine mRNA-1893 in adults aged 18 through 65 years and living in endemic and non-endemic flavivirus areas 
 NCT03014089 Phase I mRNA-1325 ModernaTX, Inc. Safety and immunogenicity of mRNA 1325 zika vaccine in healthy adults in a non-endemic Zika region 
 NCT04064905 Phase I mRNA-1893 ModernaTX, Inc. Safety, tolerability, and immunogenicity of Zika vaccine mRNA-1893 in healthy flavivirus seropositive and seronegative adults 
HIV-1 NCT03547245 Phase I mRNA-1644 International AIDS Vaccine Initiative Safety and immunogenicity of eOD-GT8 60mer vaccine, adjuvanted in HIV-uninfected, healthy adult volunteers 
RABV NCT04062669 Phase I GSK3903133A GlaxoSmithKline Safety and immunogenicity of GSK's rabies G SAM (CNE) vaccine [GSK3903133A] in healthy adults 
 NCT03713086 Phase I CV7202 CureVac Safety, reactogenicity and immunogenicity of one or two administrations of candidate rabies mRNA vaccine CV7202 in healthy adult subjects 
 NCT02241135 Phase I Nadorameran CureVac Safety and immunogenicity trial of an investigational RNActive® Rabies vaccine (CV7201) in healthy adults 
CMV NCT04232280 Phase II mRNA-1647 ModernaTX, Inc. Safety and immunogenicity of cytomegalovirus vaccine mRNA-1647 in healthy adults 
 NCT03382405 Phase I mRNA-1443, mRNA-1647 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of cytomegalovirus vaccines mRNA-1647 and mRNA-1443 when administered to healthy adults 
VZV NCT05871541 Phase I JCXH-105 Immorna Biotherapeutics, Inc. Safety and immunogenicity of a Herpes Zoster (HZ) v accine, JCXH-105, in healthy subjects 50 to 69 years of age. 
NiV NCT05398796 Phase I mRNA-1215 NIAID Safety, tolerability and immunogenicity of a nipah virus (NiV) mRNA vaccine, mRNA-1215, in healthy adults 
PfCSP NCT05581641 Phase I BNT165b1 BioNTech SE Safety, tolerability and immunogenicity of an investigational RNA-based vaccine for active immunization against malaria 
CHIKV NCT03325075 Phase I mRNA-1388 ModernaTX, Inc. Safety and immunogenicity of mRNA-1388 in healthy adults in a non-endemic Chikungunya region 
PathogenClinicalTrials.Gov IDLatest R&D statusVaccine nameSponsorMain purpose
COVID-19 NCT05375838 Phase I/II Elasomeran, mRNA-1073,mRNA-1010 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1073 (SARS-CoV-2 and Influenza Vaccine). Compared with co-administered mRNA-1010 (Influenza) in healthy adults 18–75 years of age 
 NCT06354998 Phase III Elasomeran, mRNA-1273.815 ModernaTX, Inc. Randomized, observer-blind, active-controlled of mRNA-1273.815 COVID-19 vaccine in previously vaccinated adults 
 NCT05137236 Phase II Elasomeran, mRNA-1283, mRNA-1283.211, mRNA-1283.529 ModernaTX, Inc. Immunogenicity and safety of mRNA-1283 vaccine boosters for SARS-CoV-2 
 NCT04927065 Phase II/III Elasomeran, elasomeran + imelasomeran, mRNA-1273.213, mRNA-1273.815, mRNA-1273.231, mRNA-1273.211, mRNA-1273.529, elasomeran + davesomeran, mRNA-1273.617.2 ModernaTX, Inc. Immunogenicity and safety of mRNA vaccine boosters for SARS-CoV-2 variants 
 NCT04796896 Phase II/III Elasomeran, elasomeran, imelasomeran ModernaTX, Inc. Safety, tolerability, reactogenicity, and effectiveness of mRNA-1273 SARS-CoV-2 vaccine in healthy children 6 months to less than 12 years of age 
 NCT04649151 Phase II/III Elasomeran, elasomeran + davesomeran ModernaTX, Inc. Safety, reactogenicity, and effectiveness of mRNA-1273 SARS-CoV-2 vaccine in healthy adolescents 12 to < 18 years of age 
 NCT04380701 Phase I/II BNT162a1, abdavomeran, tozinameran, pidacmeran BioNTech SE Safety and immunogenicity of four prophylactic SARS-CoV-2 RNA vaccines against COVID-19 using different dosing regimens in healthy and immunocompromised adults 
 NCT05004181 Phase II BNT162b2 (B.1.617.2), BNT162b2 (B.1.1.7), BNT162b2 (B.1.1.7 + B.1.617.2) BioNTech SE A phase II trial to evaluate the safety and immunogenicity of SARS-CoV-2 monovalent and multivalent RNA-based vaccines in healthy subjects 
 NCT04649021 Phase II Tozinameran BioNTech SE Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (BNT162b2) in Chinese healthy population: A phase ii, randomized, placebo-controlled, observer-blinded study 
 NCT04537949 Phase I/II Ganulameran BioNTech SE Safety and immunogenicity of a prophylactic SARS-CoV-2 RNA vaccine (BNT162b3) against COVID-19 using different dosing regimens in healthy adults 
 NCT04523571 Phase I Abdavomeran BioNTech SE Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (BNT162b1) in Chinese healthy subjects: A phase i, randomized, placebo-controlled, observer-blind study 
 NCT04368728 Phase II/III BNT162b2s01, abdavomeran, tozinameran BioNTech SE, Pfizer Safety, tolerability, immunogenicity, and efficacy of SARS-CoV-2 RNA vaccine candidates against COVID-19 in healthy individuals 
 NCT04955626 Phase III Riltozinameran, tozinameran + riltozinameran, tozinameran BioNTech SE, Pfizer Additional dose(s) of BNT162b2 in healthy individuals previously vaccinated with BNT162b2 
 NCT06178991 Phase III PF-07926307 BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of a combined modified RNA vaccine candidate against COVID-19 and influenza in healthy individuals 
 NCT05472038 Phase II/III Tozinameran + riltozinameran, tozinameran + famtozinameran,BNT162b5, tozinameran BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of BNT162b RNA-based vaccine candidates in COVID-19 vaccine-experienced healthy individuals 
 NCT06279871 Phase III ARCT-2303 Arcturus Therapeutics, Inc. Immunogenicity, reactogenicity, and safety of a saRNA COVID-19 vaccine (ARCT-2303), administered concomitantly with quadrivalent influenza vaccines, in adults 
 NCT05037097 Phase I/II ARCT-021, ARCT-165, ARCT-154 Arcturus Therapeutics, Inc. Safety, reactogenicity, and immunogenicity of 3 SARS-CoV-2 RNA vaccine candidates in adults previously vaccinated and not previously vaccinated against SARS-CoV-2 
 NCT05012943 Phase II/III ARCT-154 Vinbiocare Biotechnology Joint Stock Company Immunogenicity and efficacy of the SARS-CoV-2 saRNA vaccine ARCT-154 in adults 
 NCT05517642 Phase III Ad5-nCoV-IH, BNT162b2 CanSino Biologics Inc. Immunogenicity, efficacy and safety of inhaled (IH) viral vectored vaccine (Convidecia, CanSino) as second booster dose against emerging variants of concern (VOC) of SARS-CoV-2 to prevent breakthrough infections 
 NCT05373472 Phase II CS-2034 CanSino Biologics Inc. Safety and immunogenicity of COVID-19 mRNA vaccine in adults aged 18 years and older 
 NCT05743335 Phase I JCXH-221 Immorna Biotherapeutics, Inc. Safety and immunogenicity of JCXH-221, an MRNA-based broadly protective COVID-19 vaccine 
 NCT04863131 Phase I/II EXG-5003 Fujita Health University Safety and immunogenicity of intradermal SARS-CoV-2 vaccine EXG-5003 in healthy adults 
 NCT05960097 Phase II CV0701, CV0601 GlaxoSmithKline, CureVac Safety, reactogenicity, and immunogenicity of a booster dose of investigational COVID-19 mRNA vaccines in healthy adults who previously received a complete primary vaccination series with or without booster dose(s) 
 NCT05477186 Phase I CV0501 GlaxoSmithKline Safety and immunogenicity study of a booster dose of the investigational CV0501 mRNA COVID-19 vaccine in adults at least 18 years old 
 NCT04758962 Phase I GSK4184258A GlaxoSmithKline Safety, reactogenicity and immune response of a CoV-2 SAM (LNP) vaccine when administered intramuscularly on a 0, 1 month schedule in healthy adults 18 to 50 years of age 
 NCT04652102 Phase II/III CVnCoV CureVac Efficacy and safety of investigational SARS-CoV-2 mRNA vaccine CVnCoV in adults 18 years of age and older 
 NCT05903118 Phase I/II RBMRNA-176 Argorna Pharmaceuticals Co., LTD Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (RBMRNA-176) in healthy adults aged 18 years and older 
 NCT05897190 Phase I RBMRNA-405, CoronaVac® Argorna Pharmaceuticals Co., LTD Safety and immunogenicity of a SARS-CoV-2 mRNA vaccine (RBMRNA-405) as a booster in Chinese adults and older 
 NCT05435027 Phase I GRT-R912, GRT-R914, GRT-R918 Gritstone bio, Inc. Safety and tolerability of GRT-R912, GRT-R914, and GRT-R918 administered as prime and/or boost in healthy adult participants and people living with HIV 
 NCT05148962 Phase I GRT-R910 Gritstone bio, Inc. Safety, immunogenicity, and reactogenicity of a self-amplifying mRNA prophylactic vaccine boost against SARS-CoV-2 in previously vaccinated healthy adults 18 years and older 
 NCT05329220 Phase III ABNCoV2, tozinameran Bavarian Nordic Immunogenicity, safety, and tolerability of a single dose of ABNCoV2 vaccine in adult subjects previously vaccinated for SARS-CoV-2: A phase 3 trial in two parts-randomized, double-blind, active controlled and open-label, single-arm 
 NCT05210179 Phase II Tozinameran, Turkovac Health Institutes of Turkey Safety, efficacy, and immunogenicity of booster vaccination (TURKOVAC) against SARS-CoV-2 
 NCT05668065 Phase III CoronaVac, CoronaVac, BNT162b2 Institut Pasteur de Tunis Immunogenicity of heterologous versus homologous prime boost schedule with mRNA and inactivated COVID-19 vaccines: A single-blinded, randomized, parallel group superiority trial 
 NCT05352867 Phase II LVRNA009 AIM Vaccine Co., Ltd. Safety and immunogenicity of SARS-CoV-2 mRNA vaccine (LVRNA009) in Chinese people aged 18–59 years 
 NCT05343871 Phase IV Abdavomeran, ChAdOx1 nCoV-19, tozinameran Albert B. Sabin Vaccine Institut Immunogenicity and safety of full versus fractional dose of Pfizer/BioNTech, AstraZeneca, and Sinovac COVID-19 vaccines given as a booster dose at least 6 months after primary vaccination series or PCR-confirmed infection with SARS-CoV-2 in healthy adults 
 NCT05188677 Phase III SCB-2019, BNT162b2 Vaccine, Vaxzevria Vaccine, CoronaVac Vaccine Clover Biopharmaceuticals AUS Pty Ltd Immunogenicity and safety of adjuvanted recombinant SARS-CoV-2 trimeric S-protein subunit vaccine (SCB-2019), administered as a booster dose to adults, who previously received primary series of a COVID-19 vaccine 
 NCT04775069 Phase IV RBMRNA-405, CoronaVac® Humanity & Health Medical Group Limited Comparing the antibody response of subjects with chronic liver disease to mRNA, inactivated virus and adenovirus vector COVID-19 vaccines 
 NCT05030974 Phase IV Elasomeran, JNJ-78436735 University Medical Center Groningen Randomized multicenter study by the REnal patients COVID-19 VACcination (RECOVAC) Consortium 
 NCT04566276 Phase I/II ChulaCov19 Chulalongkorn University Safety, tolerability, and immunogenicity of the ChulaCov19 vaccine in healthy adults 
 NCT04569383 Phase I MVA-SARS-2-S, tozinameran Universitätsklinikum Hamburg-Eppendorf Safety, tolerability and immunogenicity of two ascending doses of the candidate vaccine MVA-SARS-2-S and heterologous booster vaccinations with a licensed vaccine against COVID-19 
 NCT04765436 Phase I PTX-COVID19-B Providence Therapeutics Holdings Inc. Safety, tolerability, and immunogenicity of PTX-COVID19-B vaccine in healthy seronegative adults aged 18–64 
 NCT04821674 Phase I/II DS-5670 Daiichi Sankyo Co., Ltd. Safety, immunogenicity and recommended dose of DS-5670a (COVID-19 vaccine) in Japanese healthy adults and elderly subjects 
 NCT05305573 Phase II Bimervax, tozinameran Hipra Scientific, S.L.U Immunogenicity and safety of a booster vaccination with a recombinant protein RBD fusion dimer candidate (PHH-1V) against SARS-CoV-2, in adults fully vaccinated with adenovirus vaccine against COVID-19 
 NCT05197153 Phase II Elasomeran, ChAdOx1 nCoV-19 Medigen Vaccine Biologics Corp. Safety, tolerability, and immunogenicity of heterologous booster dose with AZD1222, mRNA-1273, or MVC-COV1901 COVID-19 vaccine in adults 
 NCT05175742 Phase II PTX-COVID19-B, tozinameran Providence Therapeutics Holdings Inc. Safety, tolerability, and immunogenicity of PTX-COVID19-B compared to Pfizer-BioNTech COVID-19 vaccine in healthy seronegative adults aged 18 to 64 years 
 NCT05144139 Phase I/II SW0123 Stemirna Therapeutics Safety, immunogenicity and immune persistence of COVID-19 mRNA vaccine in healthy people aged 18 years and above 
 NCT05049226 Phase II ChAdOx1 nCoV-19, tozinameran Mahidol University Immunogenicity and safety of third dose vaccination with AstraZeneca COVID-19 vaccine or Pfizer/BioNTech COVID-19 vaccine among Thai Adults receiving two doses of Sinovac 
COVID-19, influenza nct06097273 Phase III mRNA-1083 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1083 (SARS-CoV-2 and influenza) vaccine in healthy adult participants, ≥50 years of age 
 NCT05596734 Phase I/II PF-07252220, PF-07926307, tozinameran + famtozinameran BioNTech SE, Pfizer Safety, tolerability, and immunogenicity of combined modified RNA vaccine candidates against COVID-19 and influenza in healthy individuals 
 NCT04969276 Phase II Elasomeran, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of Fluzone® high-dose quadrivalent (influenza vaccine), 2021–2022 formulation and a third dose of Moderna COVID-19 vaccine (mRNA-1273 vaccine) administered either concomitantly or singly in adults 65 years of age and older previously vaccinated with a 2-dose schedule of Moderna COVID-19 vaccine 
 NCT05970887 Phase IV Tozinameran + famtozinameran Catholic Kwandong University Immunogenicity and safety of concomitant administration of Omicron-containing bivalent COVID-19 vaccines with seasonal influenza vaccines 
COVID-19, influenza, RSV NCT05585632 Phase I mRNA-1045, elasomeran, elasomeran + imelasomeran, mRNA-1010, mRNA-1230, mRNA-1345 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of multi-component vaccines mRNA-1045 (influenza and RSV) or mRNA-1230 (influenza, RSV, and SARS-CoV-2) compared with mRNA-1010 (influenza), mRNA-1345 (RSV), and mRNA-1273.214 (SARS-CoV-2) vaccines in healthy adults 50–75 years of age 
NCT05330975 Phase III Elasomeran, elasomeran + imelasomeran, Afluria Quadrivalent, Afluria (trivalent), mRNA-1345 ModernaTX, Inc. Safety, tolerability, and immunogenicity of mRNA-1345, an mRNA vaccine targeting respiratory syncytial virus (RSV), when given alone or coadministered with a seasonal influenza vaccine or SARS-CoV-2 vaccine and when given as an open-label boost at 1 year following a primary dose in adults ≥ 50 years of age 
Covid-19, RSV NCT05886777 Phase II PF-06928316, tozinameran + famtozinameran Pfizer Safety, tolerability, and immunogenicity of combined vaccine candidate(s) against infectious respiratory illnesses, including COVID-19 and RSV, in healthy individuals 
Covid-19, HPV NCT05119855 Phase III Elasomeran, Gardasil 9 Merck Sharp & Dohme LLC Safety and immunogenicity of 2-dose regimens of 9vhpv and mRNA-1273 SARS-CoV-2 vaccines where the first dose of each vaccine are given concomitantly in boys and girls 9 to 11 Years of age 
COVID-19, S. pneumoniae NCT05158140 Phase III mRNA-1273, V110, V114 Merck Sharp & Dohme LLC Safety, tolerability, and immunogenicity of the concomitant administration of either 23-valent pneumococcal polysaccharide vaccine or 15-valent pneumococcal conjugate vaccine with a booster dose of SARS-CoV-2 mRNA vaccine in healthy adults 50 years of age or older. 
 NCT04887948 Phase III 20vPnC Pfizer Safety and immunogenicity of 20 valent pneumococcal conjugate vaccine when coadministered with a booster dose of BNT162b2 in adults 65 Years of age and older 
Influenza NCT05540522 Phase III PF-07252220 Pfizer Efficacy, safety, tolerability, and immunogenicity of a modified RNA vaccine against influenza compared to licensed inactivated influenza vaccine in healthy adults 18 years of age or older 
 NCT05227001 Phase I PF-07836391, PF-07836395, PF-07836396, PF-07836394, PF-07852352, PF-07867246 Pfizer Safety, tolerability, and immunogenicity of saRNA vaccine preparations against influenza in healthy individuals 
 NCT03076385 Phase I VAL-506440 ModernaTX, Inc. Safety and immunogenicity of H10N8 antigen mRNA in healthy adult subjects 
 NCT05827978 Phase III mRNA-1010, Fluarix Quadrivalent (FLU D-QIV) ModernaTX, Inc. Immunogenicity, reactogenicity and safety of mRNA-1010 seasonal influenza vaccine in adults 18 years and older 
 NCT05333289 Phase I/II mRNA-1030, mRNA-1010, mRNA-1020 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1020 and mRNA-1030 candidate seasonal influenza vaccines in healthy adults 
 NCT03345043 Phase I VAL-339851 ModernaTX, Inc. Safety and immunogenicity of VAL-339851 in healthy subjects 
 NCT05566639 Phase III mRNA-1010 ModernaTX, Inc. Safety and efficacy of mRNA-1010 candidate seasonal influenza vaccine in adults 50 years and older 
Influenza NCT05566639 Phase III mRNA-1010 ModernaTX, Inc. Safety and efficacy of mRNA-1010 candidate seasonal influenza vaccine in adults 50 years and older 
 NCT05972174 Phase I/II mRNA-1018 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1018 pandemic influenza candidate vaccines in healthy adults ≥ 18 years of age 
 NCT05827068 Phase I/II mRNA-1012.1, mRNA-1010.3, mRNA-1011.2, mRNA-1010, mRNA-1011.1, mRNA-1010.2 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1011.1, mRNA-1011.2, and mRNA-1012.1 candidate seasonal influenza vaccines in healthy adults 50 to 75 years of age 
 NCT05868382 Phase II mRNA-1010.4, mRNA-1010, mRNA-1010.6 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mrna vaccine candidate variations in healthy adults 18 to 49 years of age 
 NCT06028347 Phase I SQ012 Seqirus Safety, reactogenicity and immunogenicity of an investigational self-amplifying MRNA influenza vaccine in healthy adults 
 NCT05829356 Phase I H3 mRNA/LNP Vaccine (sanofi), Flublok Quadrivalent Sanofi Pasteur, a Sanofi Company Early safety data reviews to assess safety and immunogenicity of one monovalent modified influenza mRNA vaccine encapsulated in LNP, in adults aged 18 to 49 years and 60 years and above. 
 NCT05650554 Phase I/II Fluzone Quadrivalent, MRT5413, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5413 in adults aged 18 Years and older 
 NCT05553301 Phase I/II MRT5407, Fluzone Quadrivalent, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5407 in adults aged 18 Years and older 
 NCT05624606 Phase I/II Fluzone Quadrivalent, MRT5410, Flublok Quadrivalent, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of quadrivalent influenza mRNA vaccine MRT5410 in adults aged 18 years and older 
 NCT05426174 Phase I SP0273, Fluzone High-Dose Quadrivalent Sanofi Pasteur, a Sanofi Company Safety and immunogenicity of monovalent mRNA vaccine in adult participants 18 years of age and older 
 NCT03814720 Phase I VRC-FLUNPF099-00-VP NIAID Dose, safety, tolerability, and immunogenicity of an influenza H1 stabilized stem ferritin vaccine, VRCFLUNPF099-00-VP, in healthy adults 
 NCT05446740 Phase I FLU SV mRNA GlaxoSmithKline, CureVac Safety, reactogenicity and immunogenicity of an mRNA-based monovalent influenza vaccine candidate in healthy younger and older adults 
 NCT05252338 Phase I CVSQIV GlaxoSmithKline, CureVac Safety, reactogenicity and immunogenicity of the investigational seasonal quadrivalent influenza mRNA vaccine CVSQIV administered intramuscularly in healthy younger and older adults 
Influenza, RSV NCT05788237 Phase I PF-07252220, PF-06928316 Pfizer Safety, tolerability, and immunogenicity of respiratory combination vaccine candidates in older adults 
NCT06060457 Phase III Fluzone High-Dose Quadrivalent, mRNA-1345 ModernaTX, Inc. Safety, tolerability, and immunogenicity of mRNA-1345, an mRNA vaccine targeting respiratory syncytial virus, when coadministered with a high-dose, quadrivalent seasonal influenza vaccine in adults ≥ 65 years of age 
HPIV, hMPV NCT04144348 Phase I mRNA-1653 ModernaTX, Inc. Safety and immunogenicity of mRNA-1653, a combined human metapneumovirus (hMPV) and parainfluenza virus type 3 (PIV3) vaccine when administered to adults, and to children 12 to 59 months of age with serologic evidence of prior exposure 
 NCT03392389 Phase I mRNA-1653 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of mRNA-1653, a combined human metapneumovirus and human parainfluenza virus type 3 vaccine, when administered to healthy adults 
ZIKV NCT04917861 Phase II mRNA-1893 ModernaTX, Inc. Safety, tolerability, and immunogenicity of Zika vaccine mRNA-1893 in adults aged 18 through 65 years and living in endemic and non-endemic flavivirus areas 
 NCT03014089 Phase I mRNA-1325 ModernaTX, Inc. Safety and immunogenicity of mRNA 1325 zika vaccine in healthy adults in a non-endemic Zika region 
 NCT04064905 Phase I mRNA-1893 ModernaTX, Inc. Safety, tolerability, and immunogenicity of Zika vaccine mRNA-1893 in healthy flavivirus seropositive and seronegative adults 
HIV-1 NCT03547245 Phase I mRNA-1644 International AIDS Vaccine Initiative Safety and immunogenicity of eOD-GT8 60mer vaccine, adjuvanted in HIV-uninfected, healthy adult volunteers 
RABV NCT04062669 Phase I GSK3903133A GlaxoSmithKline Safety and immunogenicity of GSK's rabies G SAM (CNE) vaccine [GSK3903133A] in healthy adults 
 NCT03713086 Phase I CV7202 CureVac Safety, reactogenicity and immunogenicity of one or two administrations of candidate rabies mRNA vaccine CV7202 in healthy adult subjects 
 NCT02241135 Phase I Nadorameran CureVac Safety and immunogenicity trial of an investigational RNActive® Rabies vaccine (CV7201) in healthy adults 
CMV NCT04232280 Phase II mRNA-1647 ModernaTX, Inc. Safety and immunogenicity of cytomegalovirus vaccine mRNA-1647 in healthy adults 
 NCT03382405 Phase I mRNA-1443, mRNA-1647 ModernaTX, Inc. Safety, reactogenicity, and immunogenicity of cytomegalovirus vaccines mRNA-1647 and mRNA-1443 when administered to healthy adults 
VZV NCT05871541 Phase I JCXH-105 Immorna Biotherapeutics, Inc. Safety and immunogenicity of a Herpes Zoster (HZ) v accine, JCXH-105, in healthy subjects 50 to 69 years of age. 
NiV NCT05398796 Phase I mRNA-1215 NIAID Safety, tolerability and immunogenicity of a nipah virus (NiV) mRNA vaccine, mRNA-1215, in healthy adults 
PfCSP NCT05581641 Phase I BNT165b1 BioNTech SE Safety, tolerability and immunogenicity of an investigational RNA-based vaccine for active immunization against malaria 
CHIKV NCT03325075 Phase I mRNA-1388 ModernaTX, Inc. Safety and immunogenicity of mRNA-1388 in healthy adults in a non-endemic Chikungunya region 

The data in Table 2 were obtained from ClinicalTrials.gov.

SARS-CoV-2

mRNA-based COVID-19 vaccines represent the most extensively explored mRNA vaccine platform to date. Comirnaty® (BNT162b2) was the first mRNA vaccine authorized for emergency use by the FDA on December 11, 2020, and by the EMA on December 21, 2020 [78]. Shortly thereafter, Moderna’s mRNA-1273 vaccine also received approval. Both vaccines encode the full-length spike protein of SARS-CoV-2 and demonstrated 95.0% and 94.1% efficacy, respectively, in Phase III clinical trials [79]. To combat viral mutations, new vaccine formulations continue to be developed. For example, CSPC Pharmaceutical Group Ltd. developed SYS6006, an mRNA-based vaccine encoding the spike protein sequence with key mutations from Delta, Omicron BA.4, BA.5, and BF.7 strains, which received emergency use authorization (EUA) in China [80-82]. Moderna’s mRNA-1283, which encodes only the RBD and N-terminal domain of the spike protein, exhibited enhanced immune responses against the Omicron variant BA.4/BA.5 and the original virus strains compared to mRNA -1273.222 in Phase III clinical trials [83]. Additionally, mRNA-1283 exhibits improved stability at standard refrigeration temperatures (2–8℃), simplifying storage and distribution [84].

Combination vaccines represent a promising next-generation strategy. Moderna’s mRNA-1083, which integrates the influenza vaccine mRNA-1010 with the COVID-19 vaccine mRNA-1283, is currently in Phase III trials after demonstrating comparable or superior efficacy to licensed quadrivalent influenza vaccines and mRNA-1273 in earlier trials [85,86]. Beyond conventional mRNA vaccines, saRNA vaccines are under investigation. Arcturus Therapeutics' ARCT-154, an saRNA-based COVID-19 vaccine, demonstrated broader efficacy, longer-lasting immune protection, and reduced antigen dose requirements compared with BNT162b2, marking the first published clinical evidence of saRNA vaccine efficacy [87,88].

Clinical trials have also assessed the safety and immunogenicity of COVID-19 mRNA vaccines in special populations. Studies indicate that these vaccines elicited strong immune responses with acceptable tolerability in both elderly and pediatric populations [89-91]. Moreover, a six-month follow-up study of individuals aged 12 and older who received two doses of BNT162b2 reported an efficacy rate exceeding 86% and a favorable long-term safety profile [92]. These findings provide crucial support for broad vaccine deployment in diverse age groups.

Influenza

The segmented RNA genome of influenza viruses allows for the emergence of multiple subtypes and frequent antigenic drift, posing challenges for vaccine development. Currently, at least 18 distinct influenza A virus subtypes are circulating globally [93]. Multivalent vaccines encoding antigens for multiple subtypes offer a promising solution, which is difficult with conventional vaccines technology but feasible with RNA-based vaccine platforms.

Moderna’s mRNA-1010, a quadrivalent seasonal influenza vaccine encoding the full-length hemagglutinins (HA) of four influenza strains (A/H1N1, A/H3N2, B/Victoria, and B/Yamagata). In Phase I/II clinical trials (NCT04956575), single doses of mRNA-1010 induced hemagglutination inhibition (HAI) titers higher than those from standard inactivated vaccines against influenza A strains, while producing comparable HAI titers for influenza B strains [94,95]. However, the results of Phase III clinical trial were less favorable. mRNA-1010 was associated with higher adverse event rates compared with traditional vaccines, similar immunogenicity against influenza A, and inferior immunogenicity against influenza B [96]. These findings highlight the need for further optimization of mRNA vaccine platforms or antigen design to enhance efficacy and safety. Beyond conventional mRNA vaccines, Pfizer has developed saRNA-based influenza vaccine candidates. A Phase I clinical trial has been completed, though results have not yet been disclosed (NCT05227001, data from https://clinicaltrials.gov).

Respiratory syncytial virus

RSV is a single-stranded, negative-sense RNA virus and the leading cause of bronchitis and pneumonia in infants, young children, the elderly, and immunocompromised individuals. On May 31, 2024, the FDA approved Moderna’s mRNA-1345, encoding a stabilized prefusion F glycoprotein, marking the first RSV mRNA vaccine for older adults (≥60 years) [97], Sanofi’ SP0256, an RSV mRNA vaccine for older adults, showed promising Phase I/II clinical trials results and is currently in a Phase IIb trial in combination with human metapneumovirus (hMPV) (NCT06134648, NCT06237296). Sanofi also plans to develop a trivalent mRNA vaccine targeting RSV, hMPV, and parainfluenza virus [98].

While RSV mRNA vaccines have demonstrated preventive efficacy, safety concerns remain. The most common adverse reactions (ARs) include injection site pain, fatigue and myalgia (systemic), which are generally mild to moderate and transient [99]. However, in July 2024, the FDA placed an emergency hold on the Phase I clinical trial of mRNA-1365-P101 due to a potential safety signal [100]. Specifically, an imbalance in severe lower respiratory tract infections caused by RSV was observed, with a higher incidence in the vaccine group than in controls. The underlying cause remains unclear, highlighting the need for continued safety evaluations as mRNA vaccines become more widely used. To date, no broad-spectrum universal vaccine has been successfully developed for all age groups.

HIV

Developing an HIV vaccine has posed a longstanding challenge. Despite decades of research, numerous HIV vaccine candidates have failed in Phase III trials, and no vaccine has been approved for human use [101]. Moderna is currently conducting Phase I clinical studies to evaluate the safety and immunogenicity of the eOD-GT8 60mer mRNA vaccine (mRNA-1644) and the Core-g28v2 60mer mRNA vaccine (mRNA-1644v2-Core) in HIV-1 uninfected adults (NCT05001373). This trial aims to assess the ability of mRNA-based vaccines to elicit specific B-cell responses and drive their early maturation toward generating broadly neutralizing antibodies (bnAbs) [102]. Additionally, Moderna has launched another Phase I trial for the mRNA-1574 vaccine candidate, which is designed to encode native-like HIV trimers. The main endpoints of this trial are to evaluate the safety profile and immunogenicity of this experimental vaccine, with a specific focus on immune responses targeting HIV trimers (NCT05217641) [103]. As of now, results from the above trials have not been disclosed.

Other pathogens

mRNA vaccines are also being explored for various other viral infections. Moderna developed mRNA-1325 (NCT03014089) and mRNA-1893 (NCT04064905), both encoding the Zika premembrane (prM) and envelope (E) proteins. Phase I clinical trials demonstrated that mRNA-1893 elicited stronger serum-neutralizing antibody responses and better tolerability than mRNA-1325 after two doses [104,105]. For cytomegalovirus (CMV), Moderna’s Mrna-1647, comprising six mRNA sequences encoding the CMV pentamer complex and glycoprotein B (gB) antigens, demonstrated potent and durable humoral responses in Phase I/II trials, with superior neutralizing activity and antibody-dependent cellular cytotoxicity compared to the traditional gB/MF59 vaccine [106,107]. Currently, this vaccine is undergoing Phase III trials. In malaria research, BioNTech’s BNT165b1, an mRNA vaccine targeting Plasmodium falciparum circumsporozoite protein (PfCSP), entered Phase I clinical trials in 2022 (NCT05581641), representing a step toward effective malaria vaccines [108].

Terminated projects

Despite significant progress, several mRNA vaccine projects have faced challenges, underscoring the need for continuous optimization in vaccine design. For example, the early Zika virus mRNA vaccine (mRNA-1325) revealed that single amino acid differences disrupted virus-like particle formation, reducing immunogenicity and limiting efficacy [105]. Similarly, CureVac’s COVID-19 vaccine (CVnCoV) exhibited only 47% efficacy in Phase III trials, largely due to its lack of nucleoside modifications, which heightened innate immune activation and diminished potency [109]. In contrast, vaccines from Pfizer-BioNTech and Moderna, incorporating modified nucleosides, demonstrated significantly higher efficacy. Additionally, CureVac’s rabies vaccine (CV7201), which used protamine as a delivery system, showed route-dependent efficacy and caused adverse reactions in 78% of Phase I participants [110]. Development shifted to CV7202, which employed LNPs for improved delivery efficiency and reduced reactogenicity [111]. As viral variants continue to emerge, single-epitope vaccines may lack broad-spectrum protection. Future designs should consider multivalent strategies and optimize mRNA sequences, delivery systems, and adaptability to evolving pathogens to enhance vaccine efficacy and safety.

Stability and storage

mRNA-LNP vaccines, such as mRNA-1273 and BNT162b2, require frozen storage at −20℃ and −60℃ to −80℃, respectively, posing logistical challenges in resource-limited regions [112]. Lyophilization (freeze-drying) offers a promising solution by enhancing stability and shelf-life, potentially enabling ambient temperature storage [113]. However, the process must preserve key characteristics, such as particle size, polydispersity index, encapsulation efficiency, mRNA integrity, and immunogenicity [114]. Recent studies have demonstrated the feasibility of lyophilizing mRNA-LNP formulations. For example, Muramatsu et al. showed that mRNA-LNPs formulated in 5 mM Tris buffer (pH 8) with 10% sucrose and 10% maltose maintained their stability at room temperature for 12 weeks and at 4℃ for 24 weeks [115]. Similarly, Ai et al. described a lyophilized Omicron mRNA vaccine that remained stable at 25℃ for over six months, retaining robust humoral and cellular immune responses [116]. Moderna’s CMV vaccine (mRNA-1647) is undergoing Phase III clinical trials in healthy participants aged 16 to 40 years (NCT05085366) [117]. This lyophilized formulation has demonstrated stability at 5℃ for up to 18 months. Beyond lyophilization, alternative drying techniques, such as spray drying, spray freeze drying, thin-film freeze-drying, and supercritical spray drying, are being explored to optimize stability and vaccine performance [118]. Further research is needed to refine these approaches for large-scale implementation.

Extrahepatic tissues delivery

LNP-mRNA systems exhibit an inherent liver affinity, mainly due to ApoE-mediated uptake by hepatocytes, especially Kupffer cells and hepatic sinusoidal endothelial cells, making targeted delivery to extrahepatic tissues a significant challenge [119,120].

Several strategies are being explored to enhance selective organ targeting (SORT) [121]. Lipid modifications, LNPs synthesized with 5A2-SC8 lipids, combined with the addition of the cationic lipid DOTAP, can reduce ApoE binding and enhance targeting to other tissues, such as the lungs and spleen [122]. Adjusting the surface charge of LNPs is another effective approach to reduce liver uptake [123]. Moreover, surface modification of LNPs, such as coupling targeted molecules (like antibodies, peptides, or polysaccharides) to the LNP surface, represents another promising method for improving extrahepatic tissues delivery efficiency [124]. The choice of administration route also plays a crucial role in delivery efficiency, with nasal and intradermal administration enabling direct access to certain tissues. Furthermore, studies indicate that LNP size influences biodistribution, with nanoparticles in the 20–200 nm range demonstrating enhanced uptake by dendritic cells (DCs), thereby improving immune responses [120].

While extrahepatic mRNA delivery remains an area of active research, substantial progress has been made in targeting the lungs, tumors, and central nervous system through lipid chemistry innovations and ligand-based modifications. Future advancements must focus on refining targeting accuracy, ensuring safety, and optimizing large-scale production to expand the clinical applicability of mRNA vaccines.

LNP-mRNA vaccines represent a distinct advancement over traditional vaccines, with the successful application of BNT162b2 and mRNA-1273 during COVID-19 pandemic, highlighting the unprecedented adaptability and efficacy of mRNA vaccine technology in addressing infectious diseases. A comprehensive understanding of mRNA vaccines composition and optimization strategies is essential for further advancing this transformative technology. Optimizing RNA stability and translation efficiency has been a key focus, involving modifications such as alternative RNA types, nucleotide chemical alterations, codon optimization, and refined non-coding regions. Simultaneously, advancements in LNP formulations have enhanced cellular uptake, reduced off-target effects, and improved biodistribution. However, several challenges remain that must be addressed to realize the full potential of mRNA vaccines.

The intrinsic instability of mRNA necessitates a strict cold chain, limiting accessibility in resource-limited regions where ultra-low temperature storage is often impractical. The development of lyophilized mRNA-LNPs, which enable storage at 4℃ for up to six months, represents a major breakthrough. However, the impact of lyophilization buffer composition, processing conditions, and storage parameters on LNP integrity requires further investigation. Another major challenge is LNP liver accumulation, largely due to ApoE-mediated uptake. To overcome this, extensive research has focused on organ-specific LNP technologies, including high-throughput lipid screening, formulation optimization, and antibody-modified delivery systems. A particularly promising approach is SORT technology, which incorporates charged lipids (SORT lipids) into conventional LNP formulations, enabling targeted mRNA delivery to extrahepatic tissues.

Beyond technological challenges, ensuring equitable access to mRNA vaccines remains a critical global priority. The WHO’s fair allocation mechanism for COVID-19 vaccines through COVAX highlights the need to reduce distribution disparities and ensure mRNA vaccines reach low- and middle-income countries (LMICs). To achieve this, optimizing cold chain logistics, improving vaccine thermostability, and reducing production costs are essential. Strengthening local mRNA manufacturing capabilities in LMICs could also enhance regional vaccine security and pandemic preparedness, as evidenced by WHO-led initiatives promoting technology transfer hubs for mRNA vaccine production.

Furthermore, concerns regarding immunogenicity and long-term safety demand continued investigation. While recent clinical trials have shown promising immune responses and safety profiles, variations in vaccine efficacy across different populations emphasize the need for further validation. Additionally, the precise in vivo immune mechanisms of mRNA vaccines remain incompletely understood, necessitating deeper exploration to optimize immune response durability and broad applicability across diverse LNP formulations.

Addressing these scientific, logistical, and ethical challenges is crucial for expanding mRNA vaccine accessibility worldwide. Continued technological innovation, coupled with global cooperation in vaccine allocation and infrastructure development, will be instrumental in realizing the full potential of mRNA vaccines in combating infectious diseases on a global scale.

Summary

  • mRNA vaccines possess distinct advantages, including rapid development timelines, scalable production, and the ability to encode multiple antigens, showcasing their significant potential as next-generation vaccines.

  • Lipid nanoparticles remain the leading delivery system for mRNA vaccines. Advances in lipid design, formulation optimization, and selective organ targeting strategies continue to enhance their safety, transfection efficiency, and biodistribution.

  • Modifications to mRNA structure, including nucleoside modifications, codon optimization, and adjustments to untranslated regions, significantly improve stability, translation efficiency, and immunogenicity, thereby enhancing vaccine efficacy.

  • Ongoing clinical trials for various infectious diseases, such as SARS-Cov-2, respiratory syncytial virus, influenza, Zika virus, and HIV, have yield promising immunogenicity and safe data, with several candidates advancing to next-stage development.

  • Despite significant progress, key challenges remain, including long-term safety assessment, potential immunogenicity management, cold-chain requirements, and extrahepatic delivery improvements, all of which are critical for the advancement of future mRNA vaccine formulations.

The authors declare that there are no competing interests associated with the manuscript.

Sha Li, Lu Zheng, Jingyi Zhong: Concepualization; Literature search; Data curation; Writing-original draft.

Sha Li: Conceptualization; Visualization; Writing-original draft; Writing-review & editing. Xihui Gao (Corresponding author): Supervision; Writing-review & editing.

This work was financially supported by the Shanghai Municipal Science and Technology Major Project [ZD2021CY001].

IVT

in vitro transcription

LNPs

lipid nanoparticles

SORT

selective organ targeting

TLRs

Toll-like receptors

sa-RNA

self-amplifying RNA

1
Carter
,
A.
,
Msemburi
,
W.
,
Sim
,
S.Y.
,
Gaythorpe
,
K.A.M.
,
Lambach
,
P.
,
Lindstrand
,
A
, et al.
(
2024
)
Modeling the impact of vaccination for the immunization agenda 2030: deaths averted due to vaccination against 14 pathogens in 194 countries from 2021 to 2030
.
Vaccine
42
,
S28
S37
https://doi.org/10.1016/j.vaccine.2023.07.033
2.
Luo
,
W.R.
,
Wu
,
X.M.
,
Wang
,
W.
,
Yu
,
J.L.
,
Chen
,
Q.Q.
,
Zhou
,
X.
et al.
(
2022
)
Novel coronavirus mutations: vaccine development and challenges
.
Microb. Pathog.
173
, 105828 https://doi.org/10.1016/j.micpath.2022.105828
3.
Khalid
,
K.
and
Poh
,
C.L
. (
2023
)
The promising potential of reverse vaccinology-based next-generation vaccine development over conventional vaccines against antibiotic-resistant bacteria
.
Vaccines
11
, 1264 https://doi.org/10.3390/vaccines11071264
4.
Conde
,
J.
,
Langer
,
R.
and
Rueff
,
J
. (
2023
)
mRNA therapy at the convergence of genetics and nanomedicine
.
Nat. Nanotechnol.
18
,
537
540
https://doi.org/10.1038/s41565-023-01347-w
5
Kwon
,
H.
,
Kim
,
M.
,
Seo
,
Y.
,
Moon
,
Y.S.
,
Lee
,
H.J.
,
Lee
,
K
, et al.
(
2018
)
Emergence of synthetic mRNA: in vitro synthesis of mRNA and its applications in regenerative medicine
.
Biomaterials
156
,
172
193
https://doi.org/10.1016/j.biomaterials.2017.11.034
6.
Zhang
,
H.
,
Vandesompele
,
J.
,
Braeckmans
,
K.
,
De Smedt
,
S.C.
and
Remaut
,
K
. (
2024
)
Nucleic acid degradation as barrier to gene delivery: a guide to understand and overcome nuclease activity
.
Chem. Soc. Rev.
53
,
317
360
https://doi.org/10.1039/d3cs00194f
7.
Forenzo
,
C.
and
Larsen
,
J
. (
2023
)
Complex coacervates as a promising vehicle for mRNA delivery: a comprehensive review of recent advances and challenges
.
Mol. Pharm.
20
,
4387
4403
https://doi.org/10.1021/acs.molpharmaceut.3c00439
8.
Cullis
,
P.R.
and
Felgner
,
P.L
. (
2024
)
The 60-year evolution of lipid nanoparticles for nucleic acid delivery
.
Nat. Rev. Drug Discov.
23
,
709
722
https://doi.org/10.1038/s41573-024-00977-6
9.
Lombardo
,
D.
and
Kiselev
,
M.A
. (
2022
)
Methods of liposomes preparation: formation and control factors of versatile nanocarriers for biomedical and nanomedicine application
.
Pharmaceutics
14
, 543 https://doi.org/10.3390/pharmaceutics14030543
10.
Shah
,
S.
,
Dhawan
,
V.
,
Holm
,
R.
,
Nagarsenker
,
M.S.
and
Perrie
,
Y
. (
2020
)
Liposomes: advancements and innovation in the manufacturing process
.
Adv. Drug Deliv. Rev.
154
,
102
122
https://doi.org/10.1016/j.addr.2020.07.002
11
Verbeke
,
R.
,
Cheng
,
M.H.
and
Cullis
,
P.R
. (
2025
)
A historical overview on mRNA vaccine development
.
Trends mRNA Vaccine Res
1
27
https://doi.org/10.1002/9783527838394
12.
Zhang
,
M.M.
,
Bahal
,
R.
,
Rasmussen
,
T.P.
,
Manautou
,
J.E.
and
Zhong
,
X.B
. (
2021
)
The growth of siRNA-based therapeutics: updated clinical studies
.
Biochem. Pharmacol.
189
, 114432 https://doi.org/10.1016/j.bcp.2021.114432
13.
De
,
A.
and
Ko
,
Y.T
. (
2023
)
Why mRNA-ionizable LNPs formulations are so short-lived: causes and way-out
.
Expert Opin. Drug Deliv.
20
,
175
187
https://doi.org/10.1080/17425247.2023.2162876
14.
Omata
,
D.
,
Kawahara
,
E.
,
Munakata
,
L.
,
Tanaka
,
H.
,
Akita
,
H.
,
Yoshioka
,
Y.
et al.
(
2024
)
Effect of anti-PEG antibody on immune response of mRNA-loaded lipid nanoparticles
.
Mol. Pharm.
21
,
5672
5680
https://doi.org/10.1021/acs.molpharmaceut.4c00628
15.
Haq
,
M.A.
,
Roy
,
A.K.
,
Ahmed
,
R.
,
Kuddusi
,
R.U.
,
Sinha
,
M.
,
Hossain
,
M.S.
et al.
(
2024
)
Antibody longevity and waning following COVID-19 vaccination in a 1-year longitudinal cohort in Bangladesh
.
Sci. Rep.
14
,
11467
https://doi.org/10.1038/s41598-024-61922-6
16.
Kwok
,
S.L.
,
Cheng
,
S.M.
,
Leung
,
J.N.
,
Leung
,
K.
,
Lee
,
C.K.
,
Peiris
,
J.M.
et al.
(
2022
)
Waning antibody levels after COVID-19 vaccination with mRNA comirnaty and inactivated coronavac vaccines in blood donors, Hong Kong, April 2020 to October 2021
.
Euro Surveill.
27
, 2101197 https://doi.org/10.2807/1560-7917.ES.2022.27.2.2101197
17.
Verbeke
,
R.
,
Hogan
,
M.J.
,
Loré
,
K.
and
Pardi
,
N
. (
2022
)
Innate immune mechanisms of mRNA vaccines
.
Immunity
55
,
1993
2005
https://doi.org/10.1016/j.immuni.2022.10.014
18.
Schmidt
,
C.
and
Schnierle
,
B.S
. (
2023
)
Self-amplifying RNA vaccine candidates: alternative platforms for mRNA vaccine development
.
Pathogens
12
, 138 https://doi.org/10.3390/pathogens12010138
19.
Shi
,
Y.
,
Huang
,
J.
,
Liu
,
Y.
,
Liu
,
J.
,
Guo
,
X.
,
Li
,
J.
et al.
(
2022
)
Structural and biochemical characteristics of mRNA nanoparticles determine anti-SARS-CoV-2 humoral and cellular immune responses
.
Sci. Adv.
8
, eabo1827 https://doi.org/10.1126/sciadv.abo1827
20
Yu
,
D.
,
Walker
,
L.S.K.
,
Liu
,
Z.
,
Linterman
,
M.A.
and
Li
,
Z
. (
2022
)
Targeting TFH cells in human diseases and vaccination: rationale and practice
.
Nat. Immunol.
23
,
1157
1168
https://doi.org/10.1038/s41590-022-01253-8
21
Chen
,
W.
,
Teo
,
J.M.N.
,
Yau
,
S.W.
,
Wong
,
M.Y.M.
,
Lok
,
C.N.
,
Che
,
C.M
, et al.
(
2022
)
Chronic type I interferon signaling promotes lipid-peroxidation-driven terminal CD8+ T cell exhaustion and curtails anti-PD-1 efficacy
.
Cell Rep.
41
,
111647
https://doi.org/10.1016/j.celrep.2022.111647
22.
Lokras
,
A.G.
,
Bobak
,
T.R.
,
Baghel
,
S.S.
,
Sebastiani
,
F.
and
Foged
,
C
. (
2024
)
Advances in the design and delivery of RNA vaccines for infectious diseases
.
Adv. Drug Deliv. Rev.
213
,
115419
https://doi.org/10.1016/j.addr.2024.115419
23
Pourseif
,
M.M.
,
Masoudi-Sobhanzadeh
,
Y.
,
Azari
,
E.
,
Parvizpour
,
S.
,
Barar
,
J.
,
Ansari
,
R
, et al.
(
2022
)
Self-amplifying mRNA vaccines: mode of action, design, development and optimization
.
Drug Discov. Today
27
,
103341
https://doi.org/10.1016/j.drudis.2022.103341
24.
Yang
,
C.
and
Merlin
,
D
. (
2020
)
Lipid-based drug delivery nanoplatforms for colorectal cancer therapy
.
Nanomaterials
10
, 1424 https://doi.org/10.3390/nano10071424
25.
McKay
,
P.F.
,
Hu
,
K.
,
Blakney
,
A.K.
,
Samnuan
,
K.
,
Brown
,
J.C.
,
Penn
,
R.
et al.
(
2020
)
Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice
.
Nat. Commun.
11
,
3523
https://doi.org/10.1038/s41467-020-17409-9
26.
Carpentier
,
K.S.
and
Morrison
,
T.E
. (
2018
)
Innate immune control of alphavirus infection
.
Curr. Opin. Virol.
28
,
53
60
https://doi.org/10.1016/j.coviro.2017.11.006
27.
Miyake
,
K.
,
Shibata
,
T.
,
Fukui
,
R.
,
Sato
,
R.
,
Saitoh
,
S.I.
and
Murakami
,
Y
. (
2022
)
Nucleic acid sensing by toll-like receptors in the endosomal compartment
.
Front. Immunol.
13
, 941931 https://doi.org/10.3389/fimmu.2022.941931
28.
Linares-Fernández
,
S.
,
Lacroix
,
C.
,
Exposito
,
J.Y.
and
Verrier
,
B
. (
2020
)
Tailoring mRNA vaccine to balance innate/adaptive immune response
.
Trends Mol. Med.
26
,
311
323
https://doi.org/10.1016/j.molmed.2019.10.002
29.
Loan Young
,
T.
,
Chang Wang
,
K.
,
James Varley
,
A.
and
Li
,
B
. (
2023
)
Clinical delivery of circular RNA: lessons learned from RNA drug development
.
Adv. Drug Deliv. Rev.
197
,
114826
https://doi.org/10.1016/j.addr.2023.114826
30.
Qu
,
L.
,
Yi
,
Z.
,
Shen
,
Y.
,
Lin
,
L.
,
Chen
,
F.
,
Xu
,
Y.
et al.
(
2022
)
Circular RNA vaccines against SARS-CoV-2 and emerging variants
.
Cell
185
,
1728
1744
https://doi.org/10.1016/j.cell.2022.03.044
31.
Niu
,
D.
,
Wu
,
Y.
and
Lian
,
J
. (
2023
)
Circular RNA vaccine in disease prevention and treatment
.
Signal Transduct. Target. Ther.
8
,
341
https://doi.org/10.1038/s41392-023-01561-x
32.
Chen
,
R.
,
Wang
,
S.K.
,
Belk
,
J.A.
,
Amaya
,
L.
,
Li
,
Z.
,
Cardenas
,
A.
et al.
(
2023
)
Engineering circular RNA for enhanced protein production
.
Nat. Biotechnol.
41
,
262
272
https://doi.org/10.1038/s41587-022-01393-0
33.
Wan
,
J.
,
Wang
,
Z.
,
Wang
,
L.
,
Wu
,
L.
,
Zhang
,
C.
,
Zhou
,
M.
et al.
(
2024
)
Circular RNA vaccines with long-term lymph node-targeting delivery stability after lyophilization induce potent and persistent immune responses
.
mBio
15
, e01775-23 https://doi.org/10.1128/mbio.01775-23
34.
Zhang
,
Z.
,
Fu
,
Y.
,
Ju
,
X.
,
Zhang
,
F.
,
Zhang
,
P.
and
He
,
M
. (
2024
)
Advances in engineering circular RNA vaccines
.
Pathogens
13
, 692 https://doi.org/10.3390/pathogens13080692
35.
Wang
,
J.
,
Zhu
,
H.
,
Gan
,
J.
,
Liang
,
G.
,
Li
,
L.
and
Zhao
,
Y
. (
2024
)
Engineered mRNA delivery systems for biomedical applications
.
Adv. Mater. Weinheim
36
,
2308029
https://doi.org/10.1002/adma.202308029
36.
Shen
,
G.
,
Liu
,
J.
,
Yang
,
H.
,
Xie
,
N.
and
Yang
,
Y
. (
2024
)
mRNA therapies: pioneering a new era in rare genetic disease treatment
.
J. Control. Release
369
,
696
721
https://doi.org/10.1016/j.jconrel.2024.03.056
37.
Metkar
,
M.
,
Pepin
,
C.S.
and
Moore
,
M.J
. (
2024
)
Tailor made: the art of therapeutic mRNA design
.
Nat. Rev. Drug Discov.
23
,
67
83
https://doi.org/10.1038/s41573-023-00827-x
38.
Pardi
,
N.
,
Hogan
,
M.J.
,
Porter
,
F.W.
and
Weissman
,
D
. (
2018
)
mRNA vaccines - a new era in vaccinology
.
Nat. Rev. Drug Discov.
17
,
261
279
https://doi.org/10.1038/nrd.2017.243
39.
Zogg
,
H.
,
Singh
,
R.
and
Ro
,
S
. (
2022
)
Current advances in RNA therapeutics for human diseases
.
Int. J. Mol. Sci.
23
, 2736 https://doi.org/10.3390/ijms23052736
40.
Sioud
,
M.
,
Juzeniene
,
A.
and
Sæbøe-Larssen
,
S
. (
2024
)
Exploring the impact of mRNA modifications on translation efficiency and immune tolerance to self-antigens
.
Vaccines
12
, 624 https://doi.org/10.3390/vaccines12060624
41.
Xiong
,
W.
,
Zhao
,
Y.
,
Wei
,
Z.
,
Li
,
C.
,
Zhao
,
R.
,
Ge
,
J.
et al.
(
2023
)
N1-methyladenosine formation, gene regulation, biological functions, and clinical relevance
.
Mol. Ther.
31
,
308
330
https://doi.org/10.1016/j.ymthe.2022.10.015
42.
Tuncel
,
G.
and
Kalkan
,
R
. (
2019
)
Importance of m N6-methyladenosine (m6A) RNA modification in cancer
.
Med. Oncol.
36
,
36
https://doi.org/10.1007/s12032-019-1260-6
43.
Liu
,
A.
and
Wang
,
X
. (
2022
)
The pivotal role of chemical modifications in mRNA therapeutics
.
Front. Cell Dev. Biol.
10
, 901510 https://doi.org/10.3389/fcell.2022.901510
44
D’haese
,
S.
,
Laeremans
,
T.
,
Roover
,
S.D.
,
Allard
,
S.D.
,
Vanham
,
G.
and
Aerts
,
J.L
. (
2022
)
Efficient induction of antigen-specific CD8+ T-cell responses by cationic peptide-based mRNA nanoparticles
.
Pharmaceutics
14
, 1387 https://doi.org/10.3390/pharmaceutics14071387
45.
Almeida
,
A.M.
,
Queiroz
,
J.A.
,
Sousa
,
F.
and
Sousa
,
Â
. (
2019
)
Cervical cancer and HPV infection: ongoing therapeutic research to counteract the action of E6 and E7 oncoproteins
.
Drug Discov. Today
24
,
2044
2057
https://doi.org/10.1016/j.drudis.2019.07.011
46.
Xia
,
X
. (
2021
)
Detailed dissection and critical evaluation of the Pfizer/BioNTech and moderna mRNA vaccines
.
Vaccines
9
, 734 https://doi.org/10.3390/vaccines9070734
47
Qin
,
Q.
,
Yan
,
H.
,
Gao
,
W.
,
Cao
,
R.
,
Liu
,
G.
,
Zhang
,
X
, et al.
(
2024
)
Engineered mRNAs with stable structures minimize double-stranded RNA formation and increase protein expression
.
J. Mol. Biol.
436
,
168822
https://doi.org/10.1016/j.jmb.2024.168822
48.
Inagaki
,
M.
,
Abe
,
N.
,
Li
,
Z.
,
Nakashima
,
Y.
,
Acharyya
,
S.
,
Ogawa
,
K.
et al.
(
2023
)
Cap analogs with a hydrophobic photocleavable tag enable facile purification of fully capped mRNA with various cap structures
.
Nat. Commun.
14
,
2657
https://doi.org/10.1038/s41467-023-38244-8
49.
Ma
,
Q.
,
Zhang
,
X.
,
Yang
,
J.
,
Li
,
H.
,
Hao
,
Y.
and
Feng
,
X
. (
2024
)
Optimization of the 5’ untranslated region of mRNA vaccines
.
Sci. Rep.
14
,
19845
https://doi.org/10.1038/s41598-024-70792-x
50.
Biziaev
,
N.
,
Shuvalov
,
A.
,
Salman
,
A.
,
Egorova
,
T.
,
Shuvalova
,
E.
and
Alkalaeva
,
E
. (
2024
)
The impact of mRNA poly(A) tail length on eukaryotic translation stages
.
Nucleic Acids Res.
52
,
7792
7808
https://doi.org/10.1093/nar/gkae510
51.
Kim
,
S.C.
,
Sekhon
,
S.S.
,
Shin
,
W.R.
,
Ahn
,
G.
,
Cho
,
B.K.
,
Ahn
,
J.Y.
et al.
(
2022
)
Modifications of mRNA vaccine structural elements for improving mRNA stability and translation efficiency
.
Mol. Cell. Toxicol.
18
,
1
8
https://doi.org/10.1007/s13273-021-00171-4
52.
Ye
,
Z.
,
Harmon
,
J.
,
Ni
,
W.
,
Li
,
Y.
,
Wich
,
D.
and
Xu
,
Q
. (
2023
)
The mRNA vaccine revolution: COVID-19 has launched the future of vaccinology
.
ACS Nano
17
,
15231
15253
https://doi.org/10.1021/acsnano.2c12584
53.
Castillo-Hair
,
S.M.
and
Seelig
,
G
. (
2022
)
Machine learning for designing next-generation mRNA therapeutics
.
Acc. Chem. Res.
55
,
24
34
https://doi.org/10.1021/acs.accounts.1c00621
54
Zhang
,
H.
,
Zhang
,
L.
,
Lin
,
A.
,
Xu
,
C.
,
Li
,
Z.
,
Liu
,
K.
et al.
(
2023
)
Algorithm for optimized mRNA design improves stability and immunogenicity
.
Nature
621
,
396
403
https://doi.org/10.1038/s41586-023-06127-z
55.
Jin
,
L.
,
Zhou
,
Y.
,
Zhang
,
S.
and
Chen
,
S.J
. (
2025
)
mRNA vaccine sequence and structure design and optimization: advances and challenges
.
J. Biol. Chem.
301
, 108015 https://doi.org/10.1016/j.jbc.2024.108015
56.
Guarra
,
F.
and
Colombo
,
G
. (
2023
)
Computational methods in immunology and vaccinology: design and development of antibodies and immunogens
.
J. Chem. Theory Comput.
19
,
5315
5333
https://doi.org/10.1021/acs.jctc.3c00513
57.
Ma
,
W.
,
Fu
,
X.
,
Zhao
,
T.
,
Qi
,
Y.
,
Zhang
,
S.
and
Zhao
,
Y
. (
2024
)
Development and applications of lipid hydrophilic headgroups for nucleic acid therapy
.
Biotechnol. Adv.
74
,
108395
https://doi.org/10.1016/j.biotechadv.2024.108395
58.
Patel
,
P.
,
Fetse
,
J.
,
Lin
,
C.Y.
,
Guo
,
Y.
,
Hasan
,
M.R.
,
Nakhjiri
,
M.
et al.
(
2022
)
Development of amino acid-modified biodegradable lipid nanoparticles for siRNA delivery
.
Acta Biomater.
154
,
374
384
https://doi.org/10.1016/j.actbio.2022.09.065
59.
Kimura
,
S.
,
Khalil
,
I.A.
,
Elewa
,
Y.H.A.
and
Harashima
,
H
. (
2021
)
Novel lipid combination for delivery of plasmid DNA to immune cells in the spleen
.
J. Control. Release
330
,
753
764
https://doi.org/10.1016/j.jconrel.2021.01.005
60.
Akinc
,
A.
,
Maier
,
M.A.
,
Manoharan
,
M.
,
Fitzgerald
,
K.
,
Jayaraman
,
M.
,
Barros
,
S.
et al.
(
2019
)
The onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs
.
Nat. Nanotechnol.
14
,
1084
1087
https://doi.org/10.1038/s41565-019-0591-y
61.
Kang
,
D.D.
,
Hou
,
X.
,
Wang
,
L.
,
Xue
,
Y.
,
Li
,
H.
,
Zhong
,
Y.
et al.
(
2024
)
Engineering LNPs with polysarcosine lipids for mRNA delivery
.
Bioact. Mater.
37
,
86
93
https://doi.org/10.1016/j.bioactmat.2024.03.017
62.
Nussbaum
,
J.
,
Cao
,
X.
,
Railkar
,
R.A.
,
Sachs
,
J.R.
,
Spellman
,
D.S.
,
Luk
,
J.
et al.
(
2023
)
Evaluation of a stabilized RSV pre-fusion F mRNA vaccine: preclinical studies and phase 1 clinical testing in healthy adults
.
Vaccine
41
,
6488
6501
https://doi.org/10.1016/j.vaccine.2023.05.062
63.
Oda
,
Y.
,
Kumagai
,
Y.
,
Kanai
,
M.
,
Iwama
,
Y.
,
Okura
,
I.
,
Minamida
,
T
, et al.
(
2024
)
Immunogenicity and safety of a booster dose of a self-amplifying RNA COVID-19 vaccine (ARCT-154) versus BNT162b2 mRNA COVID-19 vaccine: a double-blind, multicentre, randomised, controlled, phase 3, non-inferiority trial
.
Lancet Infect. Dis.
24
,
351
360
https://doi.org/10.1016/S1473-3099(23)00650-3
64.
Chen
,
G.L.
,
Li
,
X.F.
,
Dai
,
X.H.
,
Li
,
N.
,
Cheng
,
M.L.
,
Huang
,
Z.
et al.
(
2022
)
Safety and immunogenicity of the SARS-CoV-2 ARCoV mRNA vaccine in Chinese adults: a randomised, double-blind, placebo-controlled, phase 1 trial
.
Lancet Microbe
3
,
e193
e202
https://doi.org/10.1016/S2666-5247(21)00280-9
65.
Chaudhary
,
N.
,
Weissman
,
D.
and
Whitehead
,
K.A
. (
2021
)
mRNA vaccines for infectious diseases: principles, delivery and clinical translation
.
Nat. Rev. Drug Discov.
20
,
817
838
https://doi.org/10.1038/s41573-021-00283-5
66.
Li
,
B.
,
Jiang
,
A.Y.
,
Raji
,
I.
,
Atyeo
,
C.
,
Raimondo
,
T.M.
,
Gordon
,
A.G.R.
et al.
(
2025
)
Enhancing the immunogenicity of lipid-nanoparticle mRNA vaccines by adjuvanting the ionizable lipid and the mRNA
.
Nat. Biomed. Eng.
9
,
167
184
https://doi.org/10.1038/s41551-023-01082-6
67.
Chen
,
K.
,
Fan
,
N.
,
Huang
,
H.
,
Jiang
,
X.
,
Qin
,
S.
,
Xiao
,
W.
et al.
(
2022
)
mRNA vaccines against SARS-CoV-2 variants delivered by lipid nanoparticles based on novel ionizable lipids
.
Adv. Funct. Mater.
32
, 2204692 https://doi.org/10.1002/adfm.202204692
68.
Eygeris
,
Y.
,
Gupta
,
M.
,
Kim
,
J.
and
Sahay
,
G
. (
2022
)
Chemistry of lipid nanoparticles for RNA delivery
.
Acc. Chem. Res.
55
,
2
12
https://doi.org/10.1021/acs.accounts.1c00544
69.
Patel
,
S.
,
Ashwanikumar
,
N.
,
Robinson
,
E.
,
Xia
,
Y.
,
Mihai
,
C.
,
Griffith
,
J.P.
et al.
(
2020
)
Naturally-occurring cholesterol analogues in lipid nanoparticles induce polymorphic shape and enhance intracellular delivery of mRNA
.
Nat. Commun.
11
,
983
https://doi.org/10.1038/s41467-020-14527-2
70.
Álvarez-Benedicto
,
E.
,
Farbiak
,
L.
,
Márquez Ramírez
,
M.
,
Wang
,
X.
,
Johnson
,
L.T.
,
Mian
,
O.
et al.
(
2022
)
Optimization of phospholipid chemistry for improved lipid nanoparticle (LNP) delivery of messenger RNA (mRNA)
.
Biomater. Sci.
10
,
549
559
https://doi.org/10.1039/d1bm01454d
71.
Kiaie
,
S.H.
,
Majidi Zolbanin
,
N.
,
Ahmadi
,
A.
,
Bagherifar
,
R.
,
Valizadeh
,
H.
,
Kashanchi
,
F.
et al.
(
2022
)
Recent advances in mRNA-LNP therapeutics: immunological and pharmacological aspects
.
J. Nanobiotechnology
20
,
276
https://doi.org/10.1186/s12951-022-01478-7
72.
Kong
,
W.
,
Wei
,
Y.
,
Dong
,
Z.
,
Liu
,
W.
,
Zhao
,
J.
,
Huang
,
Y.
et al.
(
2024
)
Role of size, surface charge, and PEGylated lipids of lipid nanoparticles (LNPs) on intramuscular delivery of mRNA
.
J. Nanobiotechnology
22
,
553
https://doi.org/10.1186/s12951-024-02812-x
73.
Shimabukuro
,
T
. (
2021
)
Allergic reactions including anaphylaxis after receipt of the first dose of Pfizer-BioNTech COVID-19 vaccine - United States, December 14-23, 2020
.
Am. J. Transplant.
21
,
1332
1337
https://doi.org/10.1111/ajt.16516
74.
Sherman
,
M.R.
,
Williams
,
L.D.
,
Sobczyk
,
M.A.
,
Michaels
,
S.J.
and
Saifer
,
M.G.P
. (
2012
)
Role of the methoxy group in immune responses to mPEG-protein conjugates
.
Bioconjug. Chem.
23
,
485
499
https://doi.org/10.1021/bc200551b
75.
Fiolet
,
T.
,
Kherabi
,
Y.
,
MacDonald
,
C.J.
,
Ghosn
,
J.
and
Peiffer-Smadja
,
N
. (
2022
)
Comparing COVID-19 vaccines for their characteristics, efficacy and effectiveness against SARS-CoV-2 and variants of concern: a narrative review
.
Clin. Microbiol. Infect.
28
,
202
221
https://doi.org/10.1016/j.cmi.2021.10.005
76
Ding
,
T.
,
Fu
,
J.
,
Yang
,
M.
,
Zhang
,
Z.
,
Ma
,
Y.
,
Wu
,
E
, et al.
(
2024
)
Hydroxy polyethylene glycol: a solution to evade human pre-existing anti-PEG antibodies for efficient delivery
.
bioRxiv
2024.2010. 2021.619346
https://doi.org/10.1101/2024.10.21.619346
77.
Xiao
,
Y.
,
Lian
,
X.
,
Sun
,
Y.
,
Sung
,
Y.C.
,
Vaidya
,
A.
,
Chen
,
Z.
et al.
(
2025
)
High-density brush-shaped polymer lipids reduce anti-PEG antibody binding for repeated administration of mRNA therapeutics
.
Nat. Mater.
1
12
https://doi.org/10.1038/s41563-024-02116-3
78.
Kyriakidis
,
N.C.
,
López-Cortés
,
A.
,
González
,
E.V.
,
Grimaldos
,
A.B.
and
Prado
,
E.O
. (
2021
)
SARS-CoV-2 vaccines strategies: a comprehensive review of phase 3 candidates
.
NPJ Vaccines
6
,
28
https://doi.org/10.1038/s41541-021-00292-w
79
Bruxvoort
,
K.J.
,
Sy
,
L.S.
,
Qian
,
L.
,
Ackerson
,
B.K.
,
Luo
,
Y.
,
Lee
,
G.S.
et al.
(
2021
)
Effectiveness of mRNA-1273 against delta, mu, and other emerging variants of SARS-CoV-2: test negative case-control study
.
BMJ
375
, e068848 https://doi.org/10.1136/bmj-2021-068848
80
Jin
,
F.
,
Qiu
,
Y.
,
Wu
,
Z.
,
Wang
,
Y.H.
,
Cai
,
C.
,
Fu
,
L.
et al.
(
2024
)
Immunogenicity and safety of A SARS-CoV-2 mRNA vaccine (SYS6006) in healthy Chinese participants: a randomized, observer-blinded, placebo-controlled phase 2 clinical trial
.
Vaccine
42
,
1561
1570
https://doi.org/10.1016/j.vaccine.2024.01.098
81.
Xu
,
K.
,
Lei
,
W.
,
Kang
,
B.
,
Yang
,
H.
,
Wang
,
Y.
,
Lu
,
Y.
et al.
(
2022
)
A novel mRNA vaccine, SYS6006, against SARS-CoV-2
.
Front. Immunol.
13
, 1051576 https://doi.org/10.3389/fimmu.2022.1051576
82.
Huo
,
H.
,
Wang
,
J.
,
Li
,
C.
,
Xiao
,
S.
,
Wang
,
H.
,
Ge
,
J.
et al.
(
2024
)
Safety and immunogenicity of a SARS-CoV-2 mRNA vaccine (SYS6006) in minks, cats, blue foxes, and raccoon dogs
.
Front. Cell. Infect. Microbiol.
14
, 1468775 https://doi.org/10.3389/fcimb.2024.1468775
83.
Makhijani
,
S.
,
Elossaily
,
G.M.
,
Rojekar
,
S.
and
Ingle
,
R.G
. (
2024
)
mRNA-based vaccines - global approach, challenges, and could be a promising wayout for future pandemics
.
Pharm. Dev. Technol.
29
,
559
565
https://doi.org/10.1080/10837450.2024.2361656
84.
Yang
,
L.
,
Gong
,
L.
,
Wang
,
P.
,
Zhao
,
X.
,
Zhao
,
F.
,
Zhang
,
Z.
et al.
(
2022
)
Recent advances in lipid nanoparticles for delivery of mRNA
.
Pharmaceutics
14
, 2682 https://doi.org/10.3390/pharmaceutics14122682
85.
Wang
,
Y.
,
Wei
,
X.
,
Liu
,
Y.
,
Li
,
S.
,
Pan
,
W.
,
Dai
,
J.
et al.
(
2024
)
Towards broad-spectrum protection: the development and challenges of combined respiratory virus vaccines
.
Front. Cell. Infect. Microbiol.
14
, 1412478 https://doi.org/10.3389/fcimb.2024.1412478
86
Cheng
,
Z.
,
Ma
,
J.
and
Zhao
,
C
. (
2024
)
Advantages of broad-spectrum influenza mRNA vaccines and their impact on pulmonary influenza
.
Vaccines
12
, 1382 https://doi.org/10.3390/vaccines12121382
87.
Hồ
,
N.T.
,
Hughes
,
S.G.
,
Ta
,
V.T.
,
Phan
,
L.T.
,
Đỗ
,
Q.
,
Nguyễn
,
T.V.
et al.
(
2024
)
Safety, immunogenicity and efficacy of the self-amplifying mRNA ARCT-154 COVID-19 vaccine: pooled phase 1, 2, 3a and 3b randomized, controlled trials
.
Nat. Commun.
15
,
4081
https://doi.org/10.1038/s41467-024-47905-1
88
Oda
,
Y.
,
Kumagai
,
Y.
,
Kanai
,
M.
,
Iwama
,
Y.
,
Okura
,
I.
,
Minamida
,
T
, et al.
(
2024
)
Persistence of immune responses of a self-amplifying RNA COVID-19 vaccine (ARCT-154) versus BNT162b2
.
Lancet Infect. Dis.
24
,
341
343
https://doi.org/10.1016/S1473-3099(24)00060-4
89
Walsh
,
E.E.
,
Frenck
,
R.W.
Jr
,
Falsey
,
A.R.
,
Kitchin
,
N.
,
Absalon
,
J.
,
Gurtman
,
A
, et al.
(
2020
)
Safety and immunogenicity of two RNA-based COVID-19 vaccine candidates
.
N. Engl. J. Med.
383
,
2439
2450
https://doi.org/10.1056/NEJMoa2027906
90
Frenck
,
R.W.
,
Klein
,
N.P.
,
Kitchin
,
N.
,
Gurtman
,
A.
,
Absalon
,
J.
,
Lockhart
,
S
, et al.
(
2021
)
Safety, immunogenicity, and efficacy of the BNT162b2 COVID-19 vaccine in adolescents
.
N. Engl. J. Med.
385
,
239
250
https://doi.org/10.1056/NEJMoa2107456
91.
Shimabukuro
,
T.T.
,
Kim
,
S.Y.
,
Myers
,
T.R.
,
Moro
,
P.L.
,
Oduyebo
,
T.
,
Panagiotakopoulos
,
L.
et al.
(
2021
)
Preliminary findings of mRNA COVID-19 vaccine safety in pregnant persons
.
N. Engl. J. Med.
384
,
2273
2282
https://doi.org/10.1056/NEJMoa2104983
92
Thomas
,
S.J.
,
Moreira
,
E.D.
,
Kitchin
,
N.
,
Absalon
,
J.
,
Gurtman
,
A.
,
Lockhart
,
S
, et al.
(
2021
)
Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine through 6 months
.
N. Engl. J. Med.
385
,
1761
1773
https://doi.org/10.1056/NEJMoa2110345
93.
Krammer
,
F.
,
Smith
,
G.J.D.
,
Fouchier
,
R.A.M.
,
Peiris
,
M.
,
Kedzierska
,
K.
,
Doherty
,
P.C.
et al.
(
2018
)
Influenza
.
Nat. Rev. Dis. Primers
4
,
3
https://doi.org/10.1038/s41572-018-0002-y
94.
Lee
,
I.T.
,
Nachbagauer
,
R.
,
Ensz
,
D.
,
Schwartz
,
H.
,
Carmona
,
L.
,
Schaefers
,
K.
et al.
(
2023
)
Safety and immunogenicity of a phase 1/2 randomized clinical trial of a quadrivalent, mRNA-based seasonal influenza vaccine (mRNA-1010) in healthy adults: interim analysis
.
Nat. Commun.
14
,
3631
https://doi.org/10.1038/s41467-023-39376-7
95.
Ananworanich
,
J.
,
Lee
,
I.T.
,
Ensz
,
D.
,
Carmona
,
L.
,
Schaefers
,
K.
,
Avanesov
,
A.
et al.
(
2025
)
Safety and immunogenicity of mRNA-1010, an investigational seasonal influenza vaccine, in healthy adults: final results from a phase 1/2 randomized trial
.
J. Infect. Dis.
231
,
e113
e122
https://doi.org/10.1093/infdis/jiae329
96.
Pardi
,
N.
and
Krammer
,
F
. (
2024
)
mRNA vaccines for infectious diseases - advances, challenges and opportunities
.
Nat. Rev. Drug Discov.
23
,
838
861
https://doi.org/10.1038/s41573-024-01042-y
97
Deng
,
L.
,
Cao
,
H.
,
Li
,
G.
,
Zhou
,
K.
,
Fu
,
Z.
,
Zhong
,
J.
et al.
(
2025
)
Progress on respiratory syncytial virus vaccine development and evaluation methods
.
Vaccines
13
,
304
https://doi.org/10.3390/vaccines13030304
98.
Terstappen
,
J.
,
Hak
,
S.F.
,
Bhan
,
A.
,
Bogaert
,
D.
,
Bont
,
L.J.
,
Buchholz
,
U.J.
et al.
(
2024
)
The respiratory syncytial virus vaccine and monoclonal antibody landscape: the road to global access
.
Lancet Infect. Dis.
24
,
e747
e761
https://doi.org/10.1016/S1473-3099(24)00455-9
99
Fitz-Patrick
,
D.
,
Mihara
,
H.
,
Mills
,
A.
,
Mithani
,
R.
,
Kapoor
,
A.
,
Dhar
,
R
, et al.
(
2024
)
Safety and immunogenicity of an mRNA-based RSV vaccine in Japanese older adults aged ≥60 years: a phase 1, randomized, observer-blind, placebo-controlled trial
.
Respir. Investig.
62
,
1037
1043
https://doi.org/10.1016/j.resinv.2024.08.011
100.
Mahase
,
E
. (
2024
)
FDA pauses all infant RSV vaccine trials after rise in severe illnesses
.
BMJ
387
, q2852 https://doi.org/10.1136/bmj.q2852
101.
Matarazzo
,
L.
and
Bettencourt
,
P.J.G
. (
2023
)
mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV
.
Front. Immunol.
14
, 1172691 https://doi.org/10.3389/fimmu.2023.1172691
102.
Nkolola
,
J.P.
and
Barouch
,
D.H
. (
2024
)
Prophylactic HIV-1 vaccine trials: past, present, and future
.
Lancet HIV
11
,
e117
e124
https://doi.org/10.1016/S2352-3018(23)00264-3
103.
Zhang
,
J.M.
,
Wang
,
Y.
,
Mouton
,
M.
,
Zhang
,
J.
and
Shi
,
M
. (
2024
)
Public discourse, user reactions, and conspiracy theories on the X platform about HIV vaccines: data mining and content analysis
.
J. Med. Internet Res.
26
, e53375 https://doi.org/10.2196/53375
104.
Barbier
,
A.J.
,
Jiang
,
A.Y.
,
Zhang
,
P.
,
Wooster
,
R.
and
Anderson
,
D.G
. (
2022
)
The clinical progress of mRNA vaccines and immunotherapies
.
Nat. Biotechnol.
40
,
840
854
https://doi.org/10.1038/s41587-022-01294-2
105.
Bollman
,
B.
,
Nunna
,
N.
,
Bahl
,
K.
,
Hsiao
,
C.J.
,
Bennett
,
H.
,
Butler
,
S.
et al.
(
2023
)
An optimized messenger RNA vaccine candidate protects non-human primates from Zika virus infection
.
NPJ Vaccines
8
,
58
https://doi.org/10.1038/s41541-023-00656-4
106.
Fierro
,
C.
,
Brune
,
D.
,
Shaw
,
M.
,
Schwartz
,
H.
,
Knightly
,
C.
,
Lin
,
J.
et al.
(
2024
)
Safety and immunogenicity of a messenger RNA-based cytomegalovirus vaccine in healthy adults: results from a phase 1 randomized clinical trial
.
J. Infect. Dis.
230
,
e668
e678
https://doi.org/10.1093/infdis/jiae114
107.
Hu
,
X.
,
Karthigeyan
,
K.P.
,
Herbek
,
S.
,
Valencia
,
S.M.
,
Jenks
,
J.A.
,
Webster
,
H.
et al.
(
2024
)
Human cytomegalovirus mRNA-1647 vaccine candidate elicits potent and broad neutralization and higher antibody-dependent cellular cytotoxicity responses than the gB/MF59 vaccine
.
J. Infect. Dis.
230
,
455
466
https://doi.org/10.1093/infdis/jiad593
108.
Tsoumani
,
M.E.
,
Voyiatzaki
,
C.
,
Efstathiou
,
A.
,
Tsuboi
,
T.
and
Palacpac
,
N.M.Q
. (
2023
)
Malaria vaccines: from the past towards the mRNA vaccine era
.
Vaccines
11
, 1452 https://doi.org/10.3390/vaccines11091452
109.
Morais
,
P.
,
Adachi
,
H.
and
Yu
,
Y.T
. (
2021
)
The critical contribution of pseudouridine to mRNA COVID-19 vaccines
.
Front. Cell Dev. Biol.
9
, 789427 https://doi.org/10.3389/fcell.2021.789427
110.
Alberer
,
M.
,
Gnad-Vogt
,
U.
,
Hong
,
H.S.
,
Mehr
,
K.T.
,
Backert
,
L.
,
Finak
,
G.
et al.
(
2017
)
Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised, prospective, first-in-human phase 1 clinical trial
.
The Lancet
390
,
1511
1520
https://doi.org/10.1016/S0140-6736(17)31665-3
111.
Gote
,
V.
,
Bolla
,
P.K.
,
Kommineni
,
N.
,
Butreddy
,
A.
,
Nukala
,
P.K.
,
Palakurthi
,
S.S.
et al.
(
2023
)
A comprehensive review of mRNA vaccines
.
Int. J. Mol. Sci.
24
, 2700 https://doi.org/10.3390/ijms24032700
112.
Crommelin
,
D.J.A.
,
Anchordoquy
,
T.J.
,
Volkin
,
D.B.
,
Jiskoot
,
W.
and
Mastrobattista
,
E
. (
2021
)
Addressing the cold reality of mRNA vaccine stability
.
J. Pharm. Sci.
110
,
997
1001
https://doi.org/10.1016/j.xphs.2020.12.006
113.
Lamoot
,
A.
,
Lammens
,
J.
,
De Lombaerde
,
E.
,
Zhong
,
Z.
,
Gontsarik
,
M.
,
Chen
,
Y.
et al.
(
2023
)
Successful batch and continuous lyophilization of mRNA LNP formulations depend on cryoprotectants and ionizable lipids
.
Biomater. Sci.
11
,
4327
4334
https://doi.org/10.1039/d2bm02031a
114.
Mohammady
,
M.
,
Mohammadi
,
Y.
and
Yousefi
,
G
. (
2020
)
Freeze-drying of pharmaceutical and nutraceutical nanoparticles: the effects of formulation and technique parameters on nanoparticles characteristics
.
J. Pharm. Sci.
109
,
3235
3247
https://doi.org/10.1016/j.xphs.2020.07.015
115
Muramatsu
,
H.
,
Lam
,
K.
,
Bajusz
,
C.
,
Laczkó
,
D.
,
Karikó
,
K.
,
Schreiner
,
P
, et al.
(
2022
)
Lyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine
.
Mol. Ther.
30
,
1941
1951
https://doi.org/10.1016/j.ymthe.2022.02.001
116
Ai
,
L.
,
Li
,
Y.
,
Zhou
,
L.
,
Yao
,
W.
,
Zhang
,
H.
,
Hu
,
Z.
et al.
(
2023
)
Lyophilized mRNA-lipid nanoparticle vaccines with long-term stability and high antigenicity against SARS-CoV-2
.
Cell Discov.
9
, 9 https://doi.org/10.1038/s41421-022-00517-9
117.
Akingbola
,
A.
,
Adegbesan
,
A.
,
Adewole
,
O.
,
Adegoke
,
K.
,
Benson
,
A.E.
,
Jombo
,
P.A.
et al.
(
2025
)
The mRNA-1647 vaccine: a promising step toward the prevention of cytomegalovirus infection (CMV)
.
Hum. Vaccin. Immunother.
21
,
2450045
https://doi.org/10.1080/21645515.2025.2450045
118.
Arte
,
K.S.
,
Chen
,
M.
,
Patil
,
C.D.
,
Huang
,
Y.
,
Qu
,
L.
and
Zhou
,
Q
. (
2025
)
Recent advances in drying and development of solid formulations for stable mRNA and siRNA lipid nanoparticles
.
J. Pharm. Sci.
114
,
805
815
https://doi.org/10.1016/j.xphs.2024.12.013
119.
Chu
,
R.
,
Wang
,
Y.
,
Kong
,
J.
,
Pan
,
T.
,
Yang
,
Y.
and
He
,
J
. (
2024
)
Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders
.
J. Mater. Chem. B
12
,
4759
4784
https://doi.org/10.1039/d3tb02766j
120.
Jia
,
Y.
,
Wang
,
X.
,
Li
,
L.
,
Li
,
F.
,
Zhang
,
J.
and
Liang
,
X.J
. (
2024
)
Lipid nanoparticles optimized for targeting and release of nucleic acid
.
Adv. Mater. Weinheim
36
, e2305300 https://doi.org/10.1002/adma.202305300
121.
Cheng
,
Q.
,
Wei
,
T.
,
Farbiak
,
L.
,
Johnson
,
L.T.
,
Dilliard
,
S.A.
and
Siegwart
,
D.J
. (
2020
)
Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing
.
Nat. Nanotechnol.
15
,
313
320
https://doi.org/10.1038/s41565-020-0669-6
122.
Godbout
,
K.
and
Tremblay
,
J.P
. (
2022
)
Delivery of RNAs to specific organs by lipid nanoparticles for gene therapy
.
Pharmaceutics
14
, 2129 https://doi.org/10.3390/pharmaceutics14102129
123.
Gyanani
,
V.
and
Goswami
,
R
. (
2023
)
Key design features of lipid nanoparticles and electrostatic charge-based lipid nanoparticle targeting
.
Pharmaceutics
15
, 1184 https://doi.org/10.3390/pharmaceutics15041184
124.
Jones
,
C.H.
,
Androsavich
,
J.R.
,
So
,
N.
,
Jenkins
,
M.P.
,
MacCormack
,
D.
,
Prigodich
,
A.
et al.
(
2024
)
Breaking the mold with RNA-a “RNAissance” of life science
.
NPJ Genom. Med.
9
,
2
https://doi.org/10.1038/s41525-023-00387-4

Author notes

*

These authors contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).