1. Introduction
With the continuous progress of medical science and technology, the field of cancer treatment has ushered in unprecedented changes[1-4]. As an innovative therapeutic method, tumor immunotherapy has shown great potential in cancer treatment[5]. Compared with traditional treatment, immunotherapy activates and enhances the body's own immune system to achieve precise effects on tumors, bringing new hope and possibilities for tumor patients[6-9]. However, although tumor immunotherapy has made remarkable progress in recent years, its application still faces challenges and limitations[10]. One of the main issues is how to improve the effectiveness and specificity of treatment to maximize the inhibition of tumor growth, spread and recurrence[11-14]. In this context, mRNA vaccines have attracted much attention as a potential tumor therapy. mRNA vaccines exploit nucleic acid technology. Their principle is to guide the body cells to synthesize antigen proteins by delivering specific mRNA sequences, and then the immune system produces an immune response against the tumor antigens[15-18]. Compared with traditional vaccines, mRNA vaccines have the advantages of fast preparation, strong customization, and no need to use live viruses[19]. With the rise of tumor immunotherapy, researchers have been seeking innovative ways to improve the effectiveness and specificity of treatments to better address tumor challenges. mRNA vaccines have attracted much attention as a potential tumor therapy[20]. Their principle is to guide the body cells to synthesize specific antigen proteins encoded by mRNA sequences, and then the immune system produces an immune response against the tumor antigens[21-26]. However, the clinical use of mRNA vaccines is limited by the challenges of delivery and their lack of stability. In this context, lipid nanoparticles (LNPs), which are nanomaterials, have become the key to solving the problem of mRNA vaccine delivery[27]. LNPs, as an mRNA delivery tool, have the advantages of regulability, high efficiency and stability[28-30]. The LNP structure is made up of lipid layers that wrap the mRNA and protect it from degradation. Through specific surface modification and construction schemes, LNPs can achieve targeted delivery of mRNA and enhance its enrichment in specific cells or tumor tissues, thereby improving the therapeutic effect[31-35]. The construction scheme of LNPs involves many factors, including the selection of the lipid composition, the regulation of its particle size and surface properties, and the optimization of the nucleic acid encapsulation rate[36-40]. For example, the stability and targeting of LNPs can be adjusted by rationally designing different types of lipid components. Optimizing the nucleic acid encapsulation rate can improve the delivery efficiency and bioavailability of mRNA vaccines[41]. In addition, surface modifications can enhance the specific recognition and cellular uptake of LNPs by tumor cells using targeted ligands or polymer functionalization.
However, there are some challenges in the clinical application of mRNA vaccines, namely their delivery and lack of stability[42-45]. In recent studies, lipid nanoparticles (LNPs) emerged as an effective mRNA delivery tool. LNPs have excellent biocompatibility and delivery efficiency, can be used as carriers of mRNA vaccines to improve their stability and enhance their targeting, and have shown broad application prospects in tumor immunotherapy.
This review explores the molecular mechanism of LNPs in mRNA vaccine delivery in detail, which is expected to provide theoretical guidance for further optimizing the design and construction of LNPs. This information will enhance their effectiveness and safety in tumor immunotherapy and in further understanding the targeted delivery and controlled release mechanism of LNPs, which is helpful for solving the challenges of applying mRNA vaccines in tumor immunotherapy. This study provides a scientific basis for developing more accurate and efficient tumor treatment strategies.
2. mRNA Vaccines and Tumor Immunity
2.1. Principle and Characteristics of mRNA Vaccines in Tumor Immunotherapy
2.1.1. The Basic Working Principle of mRNA Vaccines
As an innovative tumor therapy, mRNA vaccines basically work by delivering specific mRNA sequences to stimulate the body's immune system and induce an antigenic immune response against tumors[46-50]. The vaccine carries mRNA encoding specific tumor antigens that, once injected into the body, is taken up by target cells (such as dendritic cells) and translated into antigenic proteins[51]. These proteins are recognized as exogenous within the cell by the innate immune system and activate antigen-presenting cells (APCs), such as dendritic cells. The APCs present these antigens to T cells and stimulate the T cells to produce a specific immune response against these antigens[52-56]. The activated T cells will then locate and attack tumor cells that have this specific antigen, enabling targeted tumor immunotherapy.
Lipid nanoparticles (LNPs), as nanoparticle carriers containing mRNA, play an important role in whole-body transport
[57-60]. Their superior biocompatibility and efficient intracellular release mechanism make them an ideal drug delivery tool
[61-63]. LNPs can effectively protect mRNA, improve its stability, and release mRNA inside cells to promote the absorption of therapy by the target cells (
Figure 1).
2.1.2. Characteristics and Advantages of mRNA Vaccines
mRNA vaccines have unique characteristics and advantages compared with traditional vaccines
[64-66]. Their preparation speed is fast; using modern biotechnology, only the corresponding mRNA sequence is designed based on the tumor antigen sequence, and there is no need to culture an active virus or prepare a large number of proteins. mRNA vaccines can be highly personalized and can be quickly adjusted to the specific needs of different tumor types or individuals, opening up the possibility of personalized treatment
[67-70]. In addition, because mRNA vaccines can encode specific tumor antigens, they have the potential to target specific tumor antigens, which is expected to provide customized immunotherapy for different tumor types
[71-74]. In addition, mRNA vaccine preparation is relatively simple, reducing the complexity of traditional vaccine production and improving the production efficiency (
Table 1).
2.2. Current Status and Challenges of mRNA Vaccines in Tumor Therapy
2.2.1. Existing Clinical Application Cases of mRNA Vaccines
At present, several mRNA vaccines have been clinically tested in the field of tumor therapy[75]. For example, some personalized mRNA vaccines targeting specific tumor antigens have shown some clinical efficacy, prompting the body to produce an immune response against the tumor antigen. Some clinical trials[76-80] have shown that these mRNA vaccines show some therapeutic potential in some tumor types and can stimulate the body's immune system and inhibit tumor growth and spread. However, despite some progress, mRNA vaccines still face some challenges in clinical application[81]. These include stability issues, side effects control, and the challenges of generality to different tumor types[82-84]. In addition, the results of some clinical trials have not fully confirmed their efficacy and safety, and further large-scale studies and clinical validation are needed[85]. These challenges limit the widespread use of mRNA vaccines in cancer therapy, and further research and improvement are needed to improve their efficacy and reliability for clinical use.
2.2.2. The Challenges of mRNA Vaccines
mRNA vaccines as an emerging cancer therapy face multiple challenges[86-88]. The stability of mRNA vaccines is a major concern. Because mRNA is easily degraded, its stability in the body is challenged, potentially leading to the degradation and invalidation of vaccines. Therefore, how to enhance the stability of mRNA vaccines and prolong their existence time in vivo has become an urgent problem to be solved[89]. Immune response regulation is also one of the challenges faced by mRNA vaccines in tumor therapy[90]. Overactivation of the immune system can lead to adverse reactions, such as immune-related toxicity and immunoreactive side effects[91]. Therefore, how to balance and regulate the response of the immune system to ensure that the vaccine does not trigger inappropriate inflammation or autoimmune damage when inducing immunity becomes a key issue in the application of mRNA vaccines. In addition, the versatility of mRNA vaccines across different tumor types and individuals is also a challenge[92-95]. Due to tumor heterogeneity and individual patient differences, it is difficult for a single mRNA vaccine to cover all tumor types. Therefore, it is necessary to develop more widely applicable and scalable mRNA vaccines to meet the therapeutic needs of patients with different tumors.
In summary, mRNA vaccines face many challenges in tumor therapy, such as stability, immune response regulation and versatility. Overcoming these challenges will require comprehensive and in-depth research, combined with advanced technology and multidisciplinary collaboration, to address these issues and further improve the safety and effectiveness of RNA vaccines in tumor therapy.
2.3. Tumor Immune Mechanism Induced by mRNA Vaccines
2.3.1. Immunogenicity and Immune Memory
mRNA vaccines activate the body's immune system by delivering specific mRNA sequences and inducing host cells to synthesize specific tumor antigen proteins
[96-98]. These antigenic proteins are presented to T cells by antigen-presenting cells, triggering specific immune responses and promoting the activation and proliferation of CD8+ T cells and CD4+ T cells
[99]. mRNA vaccines also contribute to the formation of immune memory, allowing the body to remember and recognize specific tumor antigens in the long term, thereby rapidly generating a specific immune response when exposed to the same antigen again (
Figure 2).
2.3.2. Immune Cells and Tumor Antigens
mRNA vaccines play an important role in tumor therapy by mobilizing immune cells to recognize and attack tumor-specific antigens[100]. These vaccines work by delivering mRNA sequences encoding tumor-specific antigens, driving antigen expression within host cells, and promoting immune system activation. Dendritic cells are key cells in the immune system that are able to take up exogenous antigens and present them to T cells to initiate specific immune responses[101-104]. mRNA vaccines activate dendritic cells to take up and present tumor-specific antigens, triggering the activation and proliferation of T cells[105]. CD8+ T cells (cytotoxic T cells) play a key role in this process. They are activated and transformed into killer effector cells that seek out and attack tumor cells that express tumor-specific antigens. On the other hand, CD4+ T helper cells provide auxiliary support, promote the activation and proliferation of CD8+ T cells, and strengthen the immune response[106-110]. In addition to T cells, NK cells also play an important role[111]. mRNA vaccines promote the activation of NK cells, which are able to directly recognize and kill tumor cells expressing tumor antigens, enhancing the immune attack against tumors.
mRNA vaccines can induce the expression of tumor-specific antigens by activating dendritic cells and triggering the activation and proliferation of CD8+ T cells, CD4+ T cells and NK cells to achieve specific immune attacks against tumors[112-114]. An in-depth understanding of this mechanism could help optimize the design of mRNA vaccines and improve their efficacy and safety in tumor immunotherapy.
2.4. Development and Future Prospects of mRNA Vaccines in Tumor Immunotherapy
mRNA vaccines, as cutting-edge tumor therapies, have shown broad development prospects[115]. In the future, mRNA vaccines are expected to play an important role in tumor treatment, especially in personalized treatment. Their flexibility and customizability enable them to be precisely designed for specific tumor antigens, providing personalized, customized treatment options for all types of tumors[116-118]. mRNA vaccines are expected to show potential advantages in preventing recurrence, treating metastatic tumors, and assisting other therapeutic methods. In addition, mRNA vaccines may become an important part of tumor immunotherapy in the future, combined with immune checkpoint inhibitors or other immunotherapies to form a diversified tumor treatment regimen.
However, mRNA vaccines still face many challenges in tumor immunotherapy[119], such as improving the stability of mRNA vaccines, enhancing the specificity and persistence of the immune response, and avoiding immune-related adverse reactions[120-122]. The key to addressing these challenges lies in further in-depth research into the design and delivery of mRNA vaccines to enhance their stability and effectiveness in vivo[123-126]. In addition, the combination of new nanotechnology, biomaterials and cutting-edge technologies such as gene editing is expected to provide more effective solutions and provide more reliable support and development for the application of mRNA vaccines in tumor therapy.
3. The Role of Lipid Nanoparticles (LNPs) in mRNA Vaccine Delivery
3.1. Structure and Characteristics of LNPs
Lipid nanoparticles (LNPs), as carriers of mRNA vaccines, play an important role in mRNA delivery
[127-130]. LNPs are usually composed of hydrophobic lipids, cholesterol, PEG-modified lipids, and ionic surfactants, and they come in different nanomedicine carrier types with different applications, as shown in
Table 1. These components form a nanoscale structure whose core is a lipid double layer made of hydrophobic lipids that envelops the mRNA vaccine
[131-134]. This structure gives LNPs excellent biocompatibility and stability, helping to protect the mRNA from degradation. In addition, the surface of LNPS is often modified with PEG, which can improve their blood circulation time and reduce their chance of being cleared by the immune system. LNPs have multiple advantages in RNA vaccine delivery
[135-137]. Their lipid bilayer structure can effectively encapsulate mRNA vaccines and protect them from degradation by the external environment, which helps to improve the stability of mRNA. LNPs can improve the biological distribution of mRNA in the body and its efficiency of cell uptake and promote the delivery of mRNA to target cells, thus enhancing the effectiveness of mRNA vaccines
[138-140]. With GeoMx™ spatial analysis, scientists were able to delve deeper into the RNA needed to build lipid nanoparticles (LNPs) to more fully understand their composition and properties (
Figure 3).
Table 2.
Induction and analysis of nanomedicine carrier types and applications.
Table 2.
Induction and analysis of nanomedicine carrier types and applications.
Nanoparticle Carrier Type |
Nanomaterial Properties |
Related Research |
Targeted Tumor |
Types of Nanomedicine |
Liposomes |
Lipid bilayer structure, high encapsulation ability |
Doxil, Onivyde |
Ovarian cancer, Pancreatic cancer |
Chemotherapeutic drug delivery |
Polymeric Nanoparticles |
Tunable release properties |
Abraxane, Genexol-PM |
Breast cancer, Gastric cancer |
Chemotherapeutic drug delivery |
Gold Nanoparticles |
Biocompatibility, surface-enhanced Raman scattering |
- |
Lung cancer, Breast cancer |
Tumor photothermal therapy |
Iron Oxide Magnetic Nanoparticles |
Magnetic properties, imaging functionality |
Ferumoxytol |
Brain tumors, Breast cancer |
Magnetic resonance imaging |
Metal-Organic Frameworks (MOFs) |
High drug-loading capacity, controlled release |
- |
Lung cancer, Colorectal cancer |
Drug delivery, Imaging |
Graphene Oxide |
Large surface area, drug-loading capability |
- |
Lung cancer, Breast cancer |
Drug delivery |
Carbon Nanotubes |
High drug-loading capacity, biocompatibility |
- |
Lung cancer, Breast cancer |
Drug delivery, Photothermal therapy |
Protein Nanoparticles |
Biocompatibility, specific targeting |
Abraxane |
Pancreatic cancer, Ovarian cancer |
Protein drug delivery |
Lipid Nanoparticles |
Biocompatibility, high drug-loading capacity |
Pfizer-BioNTech mRNA vaccine |
Breast cancer, Colorectal cancer |
mRNA vaccines |
Iron Oxide Nanoparticles |
Magnetic properties, imaging functionality |
- |
Liver cancer, Breast cancer |
Magnetic resonance imaging |
PLGA Nanoparticles |
Biodegradability, controlled release |
- |
Lung cancer, Breast cancer |
Drug delivery |
Protein Polymer Nanocomplexes |
Targeted, biocompatible |
- |
Gastric cancer, Colorectal cancer |
Protein drug delivery |
Phospholipid Nanoparticles |
Biocompatibility, stability |
- |
Gastric cancer, Liver cancer |
Drug delivery |
Silica Nanoparticles |
Tunable morphology, drug-loading capability |
- |
Liver cancer, Breast cancer |
Drug delivery |
Polymer Micelles |
High drug-loading capacity, solubility |
- |
Lung cancer, Pancreatic cancer |
Chemotherapeutic drug delivery |
Nanoemulsions |
Drug-carrying capacity, stability |
- |
Pancreatic cancer, Colorectal cancer |
Drug delivery, Treatment |
3.2. Delivery Mechanism of LNPs as mRNA Vaccine Carriers
Lipid nanoparticles (LNPs), as carriers of mRNA vaccines, play an important role in tumor therapy
[141]. Their delivery mechanism is mainly manifested in two aspects: targeted delivery and controlled release. LNPs achieve targeted delivery of mRNA vaccines through their special structural and chemical properties
[142-144]. The lipid bilayer structure of LNPs enables them to encapsulate mRNA vaccines, forming stable nanoparticles that help protect mRNA from degradation
[145]. In addition, the LNP surface can be targeted by changing the lipid composition and surface modifications
[146]. Tumor-specific surface markers can improve the affinity of LNPs to tumor tissues, promote the enrichment of LNP vectors and their supported mRNA vaccines in tumor cells, and reduce their impact on healthy tissues. LNPs have the characteristics of controlled release, which helps to improve the effect of mRNA vaccines
[147-150]. Researchers can achieve the controlled release of mRNA by regulating the lipid composition and structure of LNPs so that the mRNA vaccine can be maintained in the body for a longer time and enhance its therapeutic effect (
Figure 4). In addition, LNPs can also promote the intracellular uptake of mRNA so that mRNA vaccines can enter the cell more effectively and initiate the immune response to improve the specific attack ability of tumor cells.
As the carrier of mRNA vaccines, lipid nanoparticles (LNPs) can improve the effectiveness of mRNA vaccines in tumor therapy through targeted delivery and controlled release mechanisms
[151-153]. Their targeting and controlled release properties make them a potential tumor therapeutic delivery tool, which is expected to lead to more accurate and effective treatment strategies for tumor immunotherapy (
Figure 5).
4. Application of LNP-Assisted mRNA Vaccines in Tumor Immunotherapy
4.1. Progress of Experimental Research
The application of LNP-assisted mRNA vaccines in the field of tumor therapy has aroused extensive research interest[154]. In past studies[155-160], researchers have achieved a series of encouraging results by using LNP vectors to deliver mRNA vaccines to tumor models.
Some studies have shown that LNP vectors can effectively deliver mRNA vaccines and stimulate tumor antigen-specific immune responses in tumor mouse models[161-164]. For example, some mRNA vaccines targeting tumor-specific antigens delivered through LNP carriers can induce high levels of specific antibodies and cellular immune responses, inhibit tumor growth and prolong the survival time of mice[165]. In addition, LNP-assisted mRNA vaccines have also been shown to activate CD8+ T cells and enhance immune cell recognition and attacks on tumors, playing an important role in tumor inhibition. Some studies[166-170] have pointed out that LNP carriers can help improve the stability and intracellular uptake efficiency of mRNA vaccines, thus enhancing the biological activity and persistence of mRNA vaccines. These findings provide strong support and evidence for the application of LNP-assisted mRNA vaccines in tumor therapy[171].
However, despite these positive research advances, there are some challenges and directions for future research
[172]. The biological distribution, stability, and interaction with the immune system of LNP vectors still need to be further studied to improve their delivery efficiency and reduce any potential toxic effects
[173-175]. At the same time, more preclinical studies and clinical trials will help to fully evaluate the potential use of LNP-assisted mRNA vaccines in tumor immunotherapy and their safety and efficacy (
Figure 6).
4.3. Other Potential Application Areas
LNP auxiliary mRNA vaccines are not limited to single applications in the field of tumor immunotherapy but also show broad prospects for combined applications, especially in combination with other immunotherapies[176-180]. This combined treatment strategy is expected to improve the effectiveness of tumor therapy, enhance the immune response, and overcome the limitations of a single treatment approach[181-184]. One potential application is the combination of LNP-assisted mRNA vaccines with immune checkpoint inhibitors. Immune checkpoint inhibitors activate the body's immune system to fight tumors by removing the immunosuppression of tumor cells on T cells[185-188]. LNP auxiliary mRNA vaccines can stimulate and enhance the immune response to tumor-specific antigens[189]. The combined application of the two is expected to complement each other, improve the effect of tumor immunotherapy, and may expand their scope of applications[190]. In addition, the combination of LNP-assisted mRNA vaccines with other immunotherapies, such as CAR T cell therapy or tumor vaccines, is also attracting attention[191-194]. This combined application can work synergistically to enhance multiple attacks on tumors. For example, mRNA vaccines can induce the body to produce specific antibodies and T cell immune responses, while CAR T cell therapy works by modifying T cells to directly recognize and attack tumor cells, and the combination of the two may achieve more comprehensive and long-lasting tumor treatment effects. However, these combination treatment strategies require more in-depth research to address a number of challenges, including the optimization of treatment protocols, management of side effects, and long-term monitoring of treatment effects[195-198]. In addition, the specific mechanisms and interactions of combination therapy also need to be clarified in additional experimental and clinical studies[199].
LNP auxiliary mRNA vaccines have great potential in combination with other immunotherapies, which can provide more comprehensive and effective treatment strategies for tumor immunotherapy and provide more treatment options for patients.
5. Future Prospects and Challenges
LNPs, as carriers of mRNA vaccines, show great potential in tumor immunotherapy, but they still face a series of challenges and development directions[200-205]. The future development trend of LNP vectors in tumor immunotherapy may focus on improving their delivery efficiency and accuracy[206-210]. This includes further improving the design of LNPs and optimizing their distribution and stability in vivo to improve the delivery efficiency and antitumor effect of mRNA vaccines[211]. At the same time, according to different tumor types and individual patient differences, the development of personalized and customized LNP carriers and mRNA vaccine programs is also an important direction for future development[212-218]. LNP research in tumor immunotherapy will also focus more on safety and the management of side effects. With the promotion of LNPs in clinical applications, it is necessary to have a more in-depth understanding of their metabolic dynamics and toxic reactions in the body, the ability to engage in timely detection and remediation of potential safety risks, and be able to ensure the safety and controllability of the treatment[219-222]. In addition, LNP vectors may be combined with emerging technologies such as nanotechnology and gene editing in the future to explore a variety of new therapeutic strategies[223-226]. For example, novel nanomaterials or carrier technologies can be combined to optimize LNP delivery characteristics[227-230]. Alternatively, gene editing technology and LNP carriers can be combined to achieve accurate editing and regulation of tumor genes, bringing more possibilities to tumor treatment[231-236].
However, there are still some challenges in the future development of LNP vectors in tumor immunotherapy[237-240]. This includes improving their delivery efficiency and specificity, overcoming immune-related side effects, exploring more effective targeting strategies, and reducing costs to improve production processes. Addressing these challenges requires interdisciplinary collaboration, integration of technologies and resources, strengthening of basic research and clinical trials, and continuous improvement of regulatory policies to drive continued innovation and the development of LNP vectors in the field of tumor immunotherapy[241-248].
LNPs have potential as carriers for mRNA vaccines in tumor therapy, but there are still some challenges that need to be overcome to achieve their widespread application[249]. One of the challenges is the stability and immunogenicity of LNPs in vivo[250-252]. LNPS may suffer from protein adsorption and micellar rupture in the blood circulation, limiting their ability to effectively deliver mRNA vaccines. One solution may be to improve the surface modifications of LNPs, using a variety of modifications (e.g., PEG-ification) to improve their stability and blood circulation time and to reduce immune responses[253-255]. Another challenge is the liver enrichment of LNPs. LNPs tend to be concentrated in the liver rather than tumor tissue, which limits their precise delivery to tumors[256-260]. In response to this challenge, we can explore improving the targeting of LNPs, designing specific targeting ligands or functionalized molecules, and making them more inclined to be enriched in tumor tissues to improve the therapeutic effect[261].
In addition, the LNP preparation process, production cost and scale production are also challenges[262-265]. To solve these problems, it is necessary to optimize the preparation process, increase the yield, reduce the cost, and promote large-scale production. In addressing these challenges, interdisciplinary collaboration is essential[266]. Combining expertise in biomedical science, nanotechnology, materials science and other fields strengthens research cooperation and jointly overcomes technical problems[267]. In addition, the guidance and norms of regulatory policies should be strengthened to ensure the safety and effectiveness of LNPs in clinical applications[268].
In summary, overcoming the challenges faced by LNPs as mRNA vaccine carriers in tumor therapy requires multifaceted efforts and innovation. By continuously improving the stability, targeting and production technology level of LNPs, combined with reasonable research and development strategies, it is believed that LNPs will have broader application prospects in tumor therapy.
6. Conclusions
This study summarized the key role of LNPs as mRNA vaccine carriers in tumor immunotherapy. LNPs can promote the targeted delivery and controlled release of mRNA vaccines, stimulate the immune response and fight against tumors. The advantages of mRNA vaccines are rapid preparation, personalized customization, potential for specific tumor antigens, etc., which is expected to become an innovative means of tumor treatment, and LNP-assisted mRNA vaccines have achieved encouraging therapeutic effects in tumor models.
In the future, the development prospects of LNP-assisted mRNA vaccines in tumor therapy are broad. The potential for personalized treatment and the application of combined immunotherapy will become an important direction. However, challenges such as stability, targeting, and advancing preclinical and clinical studies still need to be addressed. Further study of LNP structure optimization, targeting strategies, and multidisciplinary cooperation are suggested approaches to improve the application effect of LNP-assisted mRNA vaccines in tumor therapy and promote their clinical transformation.
Author Contributions
L.W. analyzed the data and wrote the paper; X.L. designed the research; J.L. and J.Y. guided the research; X.LQ, S.W. collected and downloaded the data in our research. All authors reviewed, edited, and approved the final manuscript and revised it critically for important intellectual content, gave final approval of the version to be published, and agreed to be accountable for all aspects of the work.
Funding
This work was supported by the National Natural Science Foundation of China (81972829), the Scientific Research Foundation of Peking University Shenzhen Hospital (KYQD202100X), Research and Development of Intelligent Surgical Navigation and Operating System for Precise Liver Resection (2022ZLA006), Start-up Fund for Talent Researchers of Tsinghua University (10001020507), National Science and Technology Major Project of China (2017ZX100203205), 2018 Peking University-University of Michigan JI Project[2019020(PUSH)-r1], 2018 Shenzhen Science and Technology Innovation Gene Project (JCYJ201802281755 31145) and 2020 Open Fund Project of Shenzhen Huada School of Life Sciences (BGIRSZ20200003).
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
Not applicable.
Conflicts of Interest
The authors declare no conflflict of interest.
References
- Bevers S, Kooijmans SAA, Van de Velde E, et al. mRNA-LNP vaccines tuned for systemic immunization induce strong antitumor immunity by engaging splenic immune cells. Mol Ther. 2022 Sep 7;30(9):3078-3094. [CrossRef]
- Ramos da Silva J, Bitencourt Rodrigues K, Formoso Pelegrin G, et al. Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice. Sci Transl Med. 2023 Mar 8;15(686):eabn3464. [CrossRef]
- Sittplangkoon C, Alameh MG, Weissman D, et al. mRNA vaccine with unmodified uridine induces robust type I interferon-dependent anti-tumor immunity in a melanoma model. Front Immunol. 2022 Oct 14;13:983000. [CrossRef]
- Zhang R, Shao S, Piao Y, et al. Esterase-Labile Quaternium Lipidoid Enabling Improved mRNA-LNP Stability and Spleen-Selective mRNA Transfection. Adv Mater. 2023 Nov;35(46):e2303614. [CrossRef]
- Li F, Zhang XQ, Ho W, et al. mRNA lipid nanoparticle-mediated pyroptosis sensitizes immunologically cold tumors to checkpoint immunotherapy. Nat Commun. 2023 Jul 15;14(1):4223. [CrossRef]
- Liu W, Alameh MG, Yang JF, et al. Lipid Nanoparticles Delivering Constitutively Active STING mRNA to Stimulate Antitumor Immunity. Int J Mol Sci. 2022 Nov 22;23(23):14504. [CrossRef]
- Kitte R, Rabel M, Geczy R, et al. Lipid nanoparticles outperform electroporation in mRNA-based CAR T cell engineering. Mol Ther Methods Clin Dev. 2023 Oct 18;31:101139. [CrossRef]
- Qiu K, Duan X, Mao M, et al. mRNA-LNP vaccination-based immunotherapy augments CD8+ T cell responses against HPV-positive oropharyngeal cancer. NPJ Vaccines. 2023 Sep 29;8(1):144. [CrossRef]
- Golubovskaya V, Sienkiewicz J, Sun J, et al. CAR-NK Cells Generated with mRNA-LNPs Kill Tumor Target Cells In Vitro and In Vivo. Int J Mol Sci. 2023 Aug 29;24(17):13364. [CrossRef]
- Kiaie SH, Majidi Zolbanin N, Ahmadi A, et al. Recent advances in mRNA-LNP therapeutics: immunological and pharmacological aspects. J Nanobiotechnology. 2022 Jun 14;20(1):276. [CrossRef]
- Schoenmaker L, Witzigmann D, Kulkarni JA, et al. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int J Pharm. 2021 May 15;601:120586. [CrossRef]
- Ndeupen S, Qin Z, Jacobsen S, et al. The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. iScience. 2021 Dec 17;24(12):103479. [CrossRef]
- Alameh MG, Tombácz I, Bettini E, et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity. 2021 Dec 14;54(12):2877-2892.e7. [CrossRef]
- Zhang NN, Li XF, Deng YQ, et al. A Thermostable mRNA Vaccine against COVID-19. Cell. 2020 Sep 3;182(5):1271-1283.e16. [CrossRef]
- Kon E, Elia U, Peer D. Principles for designing an optimal mRNA lipid nanoparticle vaccine. Curr Opin Biotechnol. 2022 Feb;73:329-336. [CrossRef]
- Hassett KJ, Higgins J, Woods A, et al. Impact of lipid nanoparticle size on mRNA vaccine immunogenicity. J Control Release. 2021 Jul 10;335:237-246. [CrossRef]
- Zong Y, Lin Y, Wei T, et al. Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy. Adv Mater. 2023 May 17:e2303261. [CrossRef]
- Verbeke R, Hogan MJ, Loré K, et al. Innate immune mechanisms of mRNA vaccines. Immunity. 2022 Nov 8;55(11):1993-2005. [CrossRef]
- Muramatsu H, Lam K, Bajusz C, et al. Lyophilization provides long-term stability for a lipid nanoparticle-formulated, nucleoside-modified mRNA vaccine. Mol Ther. 2022 May 4;30(5):1941-1951. [CrossRef]
- Wollner CJ, Richner M, Hassert MA, et al. A Dengue Virus Serotype 1 mRNA-LNP Vaccine Elicits Protective Immune Responses. J Virol. 2021 May 24;95(12):e02482-20. [CrossRef]
- Oude Blenke E, Örnskov E, Schöneich C, et al. The Storage and In-Use Stability of mRNA Vaccines and Therapeutics: Not A Cold Case. J Pharm Sci. 2023 Feb;112(2):386-403. [CrossRef]
- Ripoll M, Bernard MC, Vaure C, et al. An imidazole modified lipid confers enhanced mRNA-LNP stability and strong immunization properties in mice and non-human primates. Biomaterials. 2022 Jul;286:121570. [CrossRef]
- Hayashi CTH, Cao Y, Clark LC, et al. mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines. 2022 Dec 1;7(1):155. [CrossRef]
- Kon E, Levy Y, Elia U, et al. A single-dose F1-based mRNA-LNP vaccine provides protection against the lethal plague bacterium. Sci Adv. 2023 Mar 10;9(10):eadg1036. [CrossRef]
- Qin Z, Bouteau A, Herbst C, et al. Pre-exposure to mRNA-LNP inhibits adaptive immune responses and alters innate immune fitness in an inheritable fashion. PLoS Pathog. 2022 Sep 2;18(9):e1010830. [CrossRef]
- McMahon M, O'Dell G, Tan J, et al. Assessment of a quadrivalent nucleoside-modified mRNA vaccine that protects against group 2 influenza viruses. Proc Natl Acad Sci U S A. 2022 Nov 8;119(45):e2206333119. [CrossRef]
- Monslow MA, Elbashir S, Sullivan NL, et al. Immunogenicity generated by mRNA vaccine encoding VZV gE antigen is comparable to adjuvanted subunit vaccine and better than live attenuated vaccine in nonhuman primates. Vaccine. 2020 Aug 10;38(36):5793-5802. [CrossRef]
- Sáez-Llorens X, Lanata C, Aranguren E, et al. Safety and immunogenicity of mRNA-LNP COVID-19 vaccine CVnCoV in Latin American adults: A phase 2 randomized study. Vaccine X. 2022 Aug;11:100189. [CrossRef]
- Hoffmann MAG, Yang Z, Huey-Tubman KE, et al. ESCRT recruitment to SARS-CoV-2 spike induces virus-like particles that improve mRNA vaccines. Cell. 2023 May 25;186(11):2380-2391.e9. [CrossRef]
- Pardi N, Hogan MJ, Naradikian MS, et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med. 2018 Jun 4;215(6):1571-1588. [CrossRef]
- Laczkó D, Hogan MJ, Toulmin SA, et al. A Single Immunization with Nucleoside-Modified mRNA Vaccines Elicits Strong Cellular and Humoral Immune Responses against SARS-CoV-2 in Mice. Immunity. 2020 Oct 13;53(4):724-732.e7. [CrossRef]
- Valentin A, Bergamaschi C, Rosati M, et al. Comparative immunogenicity of an mRNA/LNP and a DNA vaccine targeting HIV gag conserved elements in macaques. Front Immunol. 2022 Jul 22;13:945706. [CrossRef]
- Pardi N, Carreño JM, O'Dell G, et al. Development of a pentavalent broadly protective nucleoside-modified mRNA vaccine against influenza B viruses. Nat Commun. 2022 Aug 9;13(1):4677. [CrossRef]
- Aldrich C, Leroux-Roels I, Huang KB, et al. Proof-of-concept of a low-dose unmodified mRNA-based rabies vaccine formulated with lipid nanoparticles in human volunteers: A phase 1 trial. Vaccine. 2021 Feb 22;39(8):1310-1318. [CrossRef]
- Pilkington EH, Suys EJA, Trevaskis NL, et al. From influenza to COVID-19: Lipid nanoparticle mRNA vaccines at the frontiers of infectious diseases. Acta Biomater. 2021 Sep 1;131:16-40. [CrossRef]
- Naderi Sohi A, Kiani J, Arefian E, et al. Development of an mRNA-LNP Vaccine against SARS-CoV-2: Evaluation of Immune Response in Mouse and Rhesus Macaque. Vaccines (Basel). 2021 Sep 10;9(9):1007. [CrossRef]
- Pardi N, Hogan MJ, Pelc RS, et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature. 2017 Mar 9;543(7644):248-251. [CrossRef]
- Liang Q, Wang Y, Zhang S, et al. RBD trimer mRNA vaccine elicits broad and protective immune responses against SARS-CoV-2 variants. iScience. 2022 Apr 15;25(4):104043. [CrossRef]
- Bavli Y, Chen BM, Gross G, et al. Anti-PEG antibodies before and after a first dose of Comirnaty (mRNA-LNP-based SARS-CoV-2 vaccine). J Control Release. 2023 Feb;354:316-322. [CrossRef]
- Liu T, Tian Y, Zheng A, et al. Design Strategies for and Stability of mRNA-Lipid Nanoparticle COVID-19 Vaccines. Polymers (Basel). 2022 Oct 6;14(19):4195. [CrossRef]
- Qin Z, Bouteau A, Herbst C, et al. Pre-exposure to mRNA-LNP inhibits adaptive immune responses and alters innate immune fitness in an inheritable fashion. bioRxiv [Preprint]. 2022 Aug 20:2022.03.16.484616.
- He L, Sun W, Yang L, et al. A multiple-target mRNA-LNP vaccine induces protective immunity against experimental multi-serotype DENV in mice. Virol Sin. 2022 Oct;37(5):746-757. [CrossRef]
- Lederer K, Castaño D, Gómez Atria D, et al. SARS-CoV-2 mRNA Vaccines Foster Potent Antigen-Specific Germinal Center Responses Associated with Neutralizing Antibody Generation. Immunity. 2020 Dec 15;53(6):1281-1295.e5. [CrossRef]
- Ndeupen S, Qin Z, Jacobsen S, et al. The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. bioRxiv [Preprint]. 2021 Jul 23:2021.03.04.430128.
- Chen T, Zhu S, Wei N, et al. Protective Immune Responses Induced by an mRNA-LNP Vaccine Encoding prM-E Proteins against Japanese Encephalitis Virus Infection. Viruses. 2022 May 24;14(6):1121. [CrossRef]
- Guéguen C, Ben Chimol T, Briand M, et al. Evaluating how cationic lipid affects mRNA-LNP physical properties and biodistribution. Eur J Pharm Biopharm. 2023 Aug 12:S0939-6411(23)00205-9. [CrossRef]
- Hayashi CTH, Cao Y, Clark LC, et al. Author Correction: mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines. 2023 Aug 11;8(1):115. [CrossRef]
- Jansen EM, Frijlink HW, Hinrichs WL, et al. Are inhaled mRNA vaccines safe and effective? A review of preclinical studies. Expert Opin Drug Deliv. 2022 Nov;19(11):1471-1485. [CrossRef]
- Egan KP, Awasthi S, Tebaldi G, et al. A Trivalent HSV-2 gC2, gD2, gE2 Nucleoside-Modified mRNA-LNP Vaccine Provides Outstanding Protection in Mice against Genital and Non-Genital HSV-1 Infection, Comparable to the Same Antigens Derived from HSV-1. Viruses. 2023 Jun 30;15(7):1483. [CrossRef]
- Granados-Riveron JT, Aquino-Jarquin G. Engineering of the current nucleoside-modified mRNA-LNP vaccines against SARS-CoV-2. Biomed Pharmacother. 2021 Oct;142:111953. [CrossRef]
- Ndeupen S, Bouteau A, Herbst C, et al. Langerhans cells and cDC1s play redundant roles in mRNA-LNP induced protective anti-influenza and anti-SARS-CoV-2 immune responses. PLoS Pathog. 2022 Jan 24;18(1):e1010255. [CrossRef]
- Zhang Y, Li S, Chu H, et al. A novel mRNA vaccine, TGGT1_278620 mRNA-LNP, prolongs the survival time in BALB/c mice with acute toxoplasmosis. Microbiol Spectr. 2023 Dec 1:e0286623. [CrossRef]
- Zhang Y, Li D, Shen Y, et al. Immunization with a novel mRNA vaccine, TGGT1_216200 mRNA-LNP, prolongs survival time in BALB/c mice against acute toxoplasmosis. Front Immunol. 2023 Apr 14;14:1161507. [CrossRef]
- Lamoot A, Lammens J, De Lombaerde E, et al. Successful batch and continuous lyophilization of mRNA LNP formulations depend on cryoprotectants and ionizable lipids. Biomater Sci. 2023 Jun 13;11(12):4327-4334. [CrossRef]
- Ndeupen S, Bouteau A, Herbst C, et al. Langerhans cells and cDC1s play redundant roles in mRNA-LNP induced protective anti-influenza and anti-SARS-CoV-2 responses. bioRxiv [Preprint]. 2021 Aug 2:2021.08.01.454662.
- Zhang L, More KR, Ojha A, et al. Effect of mRNA-LNP components of two globally-marketed COVID-19 vaccines on efficacy and stability. NPJ Vaccines. 2023 Oct 11;8(1):156. [CrossRef]
- Ge N, Sun J, Liu Z, et al. An mRNA vaccine encoding Chikungunya virus E2-E1 protein elicits robust neutralizing antibody responses and CTL immune responses. Virol Sin. 2022 Apr;37(2):266-276. [CrossRef]
- Carter B, Huang P, Liu G, et al. A pan-variant mRNA-LNP T cell vaccine protects HLA transgenic mice from mortality after infection with SARS-CoV-2 Beta. Front Immunol. 2023 Mar 9;14:1135815. [CrossRef]
- Sáez-Llorens X, Lanata C, Aranguren E, et al. Corrigendum to "Safety and immunogenicity of mRNA-LNP COVID-19 vaccine CVnCoV in Latin American adults: A phase 2 randomized study" [Vaccine: X 11 (2022) 100189]. Vaccine X. 2023 Aug;14:100307. [CrossRef]
- Baldeon Vaca G, Meyer M, Cadete A, et al. Intranasal mRNA-LNP vaccination protects hamsters from SARS-CoV-2 infection. Sci Adv. 2023 Sep 22;9(38):eadh1655. [CrossRef]
- Patel N, Davis Z, Hofmann C, et al. Development and Characterization of an In Vitro Cell-Based Assay to Predict Potency of mRNA-LNP-Based Vaccines. Vaccines (Basel). 2023 Jul 10;11(7):1224. [CrossRef]
- Rizvi F, Lee YR, Diaz-Aragon R, et al. VEGFA mRNA-LNP promotes biliary epithelial cell-to-hepatocyte conversion in acute and chronic liver diseases and reverses steatosis and fibrosis. bioRxiv [Preprint]. 2023 Apr 18:2023.04.17.537186.
- Moyles IR, Korosec CS, Heffernan JM. Determination of significant immunological timescales from mRNA-LNP-based vaccines in humans. J Math Biol. 2023 Apr 30;86(5):86. [CrossRef]
- Li D, Zhang Y, Li S, et al. A novel Toxoplasma gondii TGGT1_316290 mRNA-LNP vaccine elicits protective immune response against toxoplasmosis in mice. Front Microbiol. 2023 Mar 21;14:1145114. [CrossRef]
- Zhang M, Sun J, Li M, et al. Modified mRNA-LNP Vaccines Confer Protection against Experimental DENV-2 Infection in Mice. Mol Ther Methods Clin Dev. 2020 Jul 21;18:702-712. [CrossRef]
- Zhang X, Jozic A, Song P, et al. mRNA vaccine against fibroblast activation protein ameliorates murine models of inflammatory arthritis. Rheumatol Immunol Res. 2023 Jul 22;4(2):90-97. [CrossRef]
- Hsu FF, Liang KH, Kumari M, et al. An efficient approach for SARS-CoV-2 monoclonal antibody production via modified mRNA-LNP immunization. Int J Pharm. 2022 Nov 5;627:122256. [CrossRef]
- Hermosilla J, Alonso-García A, Salmerón-García A, et al. Analysing the In-Use Stability of mRNA-LNP COVID-19 Vaccines Comirnaty™ (Pfizer) and Spikevax™ (Moderna): A Comparative Study of the Particulate. Vaccines (Basel). 2023 Oct 25;11(11):1635. [CrossRef]
- Egan KP, Hook LM, Naughton A, et al. An HSV-2 nucleoside-modified mRNA genital herpes vaccine containing glycoproteins gC, gD, and gE protects mice against HSV-1 genital lesions and latent infection. PLoS Pathog. 2020 Jul 27;16(7):e1008795. [CrossRef]
- Nag K, Chandra Baray J, Rahman Khan M, et al. An mRNA-based vaccine candidate against SARS-CoV-2 elicits stable immuno-response with single dose. Vaccine. 2021 Jun 23;39(28):3745-3755. [CrossRef]
- Kim D, Lai CJ, Cha I, et al. SFTSV Gn-Head mRNA vaccine confers efficient protection against lethal viral challenge. J Med Virol. 2023 Nov;95(11):e29203. [CrossRef]
- Xia H, He YR, Zhan XY, et al. Mpox virus mRNA-lipid nanoparticle vaccine candidates evoke antibody responses and drive protection against the Vaccinia virus challenge in mice. Antiviral Res. 2023 Aug;216:105668. [CrossRef]
- Hoffmann MAG, Yang Z, Huey-Tubman KE, et al. ESCRT recruitment to mRNA-encoded SARS-CoV-2 spike induces virus-like particles and enhanced antibody responses. bioRxiv [Preprint]. 2022 Dec 27:2022.12.26.521940.
- Ndeupen S, Qin Z, Igyártó BZ. Single-cell suspension preparation from murine organs following in vivo mRNA-LNP exposure. STAR Protoc. 2022 May 18;3(2):101350. [CrossRef]
- Zamani P, Mashreghi M, Rezazade Bazaz M, et al. Characterization of stability, safety and immunogenicity of the mRNA lipid nanoparticle vaccine Iribovax against COVID-19 in nonhuman primates. J Control Release. 2023 Aug;360:316-334. [CrossRef]
- Nelson CS, Jenks JA, Pardi N, et al. Human Cytomegalovirus Glycoprotein B Nucleoside-Modified mRNA Vaccine Elicits Antibody Responses with Greater Durability and Breadth than MF59-Adjuvanted gB Protein Immunization. J Virol. 2020 Apr 16;94(9):e00186-20. [CrossRef]
- Lelis F, Byk LA, Pustylnikov S, et al. Safety, immunogenicity and efficacy of an mRNA-based COVID-19 vaccine, GLB-COV2-043, in preclinical animal models. Sci Rep. 2023 Dec 1;13(1):21172. [CrossRef]
- Narayanan E, Falcone S, Elbashir SM, et al. Rational Design and In Vivo Characterization of mRNA-Encoded Broadly Neutralizing Antibody Combinations against HIV-1. Antibodies (Basel). 2022 Oct 24;11(4):67. [CrossRef]
- Mao S, Li S, Zhang Y, et al. A highly efficient needle-free-injection delivery system for mRNA-LNP vaccination against SARS-CoV-2. Nano Today. 2023 Feb;48:101730. [CrossRef]
- Chivukula S, Plitnik T, Tibbitts T, et al. Development of multivalent mRNA vaccine candidates for seasonal or pandemic influenza. NPJ Vaccines. 2021 Dec 16;6(1):153. [CrossRef]
- Rizvi F, Lee YR, Diaz-Aragon R, et al. VEGFA mRNA-LNP promotes biliary epithelial cell-to-hepatocyte conversion in acute and chronic liver diseases and reverses steatosis and fibrosis. Cell Stem Cell. 2023 Nov 20:S1934-5909(23)00392-2. [CrossRef]
- Chuang YM, Alameh MG, Abouneameh S, et al. A mosquito AgTRIO mRNA vaccine contributes to immunity against malaria. NPJ Vaccines. 2023 Jun 7;8(1):88. [CrossRef]
- Pardi N, LaBranche CC, Ferrari G, et al. Characterization of HIV-1 Nucleoside-Modified mRNA Vaccines in Rabbits and Rhesus Macaques. Mol Ther Nucleic Acids. 2019 Apr 15;15:36-47. [CrossRef]
- Gouma S, Furey C, Santos JJS, et al. Nucleoside-Modified mRNA-Based Influenza Vaccines Circumvent Problems Associated with H3N2 Vaccine Strain Egg Adaptation. J Virol. 2023 Jan 31;97(1):e0172322. [CrossRef]
- John S, Yuzhakov O, Woods A, et al. Multi-antigenic human cytomegalovirus mRNA vaccines that elicit potent humoral and cell-mediated immunity. Vaccine. 2018 Mar 14;36(12):1689-1699. [CrossRef]
- Patra T, Meyer K, Haga Y, et al. Hepatitis C virus E1 and modified E2 delivered from an mRNA vaccine induces protective immunity. NPJ Vaccines. 2023 Mar 18;8(1):42. [CrossRef]
- Zhao H, Wang TC, Li XF, et al. Long-term stability and protection efficacy of the RBD-targeting COVID-19 mRNA vaccine in nonhuman primates. Signal Transduct Target Ther. 2021 Dec 24;6(1):438. [CrossRef]
- Medjmedj A, Ngalle-Loth A, Clemençon R, et al. In Cellulo and In Vivo Comparison of Cholesterol, Beta-Sitosterol and Dioleylphosphatidylethanolamine for Lipid Nanoparticle Formulation of mRNA. Nanomaterials (Basel). 2022 Jul 17;12(14):2446. [CrossRef]
- El-Mayta R, Padilla MS, Billingsley MM, Testing the In Vitr et al.o and In Vivo Efficiency of mRNA-Lipid Nanoparticles Formulated by Microfluidic Mixing. J Vis Exp. 2023 Jan 20;(191). [CrossRef]
- Xu S, Zhang B, Yao J, et al. A new H9 influenza virus mRNA vaccine elicits robust protective immunity against infection. Vaccine. 2023 May 2;41(18):2905-2913. [CrossRef]
- Hook LM, Awasthi S, Cairns TM, et al. Antibodies to Crucial Epitopes on HSV-2 Glycoprotein D as a Guide to Dosing an mRNA Genital Herpes Vaccine. Viruses. 2022 Mar 5;14(3):540. [CrossRef]
- Ci L, Hard M, Zhang H, et al. Biodistribution of Lipid 5, mRNA, and Its Translated Protein Following Intravenous Administration of mRNA-Encapsulated Lipid Nanoparticles in Rats. Drug Metab Dispos. 2023 Jul;51(7):813-823. [CrossRef]
- Maharjan R, Hada S, Lee JE, et al. Comparative study of lipid nanoparticle-based mRNA vaccine bioprocess with machine learning and combinatorial artificial neural network-design of experiment approach. Int J Pharm. 2023 Jun 10;640:123012. [CrossRef]
- Ma Q, Li R, Guo J, et al. Immunization with a Prefusion SARS-CoV-2 Spike Protein Vaccine (RBMRNA-176) Protects against Viral Challenge in Mice and Nonhuman Primates. Vaccines (Basel). 2022 Oct 11;10(10):1698. [CrossRef]
- Ma N, Xia ZW, Zhang ZG, et al. Development of an mRNA vaccine against a panel of heterologous H1N1 seasonal influenza viruses using a consensus hemagglutinin sequence. Emerg Microbes Infect. 2023 Dec;12(1):2202278. [CrossRef]
- Cui L, Hunter MR, Sonzini S, et al. Mechanistic Studies of an Automated Lipid Nanoparticle Reveal Critical Pharmaceutical Properties Associated with Enhanced mRNA Functional Delivery In Vitro and In Vivo. Small. 2022 Mar;18(9):e2105832. [CrossRef]
- Wilhelmy C, Keil IS, Uebbing L, et al. Polysarcosine-Functionalized mRNA Lipid Nanoparticles Tailored for Immunotherapy. Pharmaceutics. 2023 Aug 1;15(8):2068. [CrossRef]
- Dézsi L, Mészáros T, Kozma G, et al. A naturally hypersensitive porcine model may help understand the mechanism of COVID-19 mRNA vaccine-induced rare (pseudo) allergic reactions: complement activation as a possible contributing factor. Geroscience. 2022 Apr;44(2):597-618. [CrossRef]
- Bai S, Yang T, Zhu C, et al. A single vaccination of nucleoside-modified Rabies mRNA vaccine induces prolonged highly protective immune responses in mice. Front Immunol. 2023 Jan 17;13:1099991. [CrossRef]
- Appelberg S, John L, Pardi N, et al. Nucleoside-Modified mRNA Vaccines Protect IFNAR-/- Mice against Crimean-Congo Hemorrhagic Fever Virus Infection. J Virol. 2022 Feb 9;96(3):e0156821. [CrossRef]
- LaTourette PC 2nd, Awasthi S, Desmond A, et al. Protection against herpes simplex virus type 2 infection in a neonatal murine model using a trivalent nucleoside-modified mRNA in lipid nanoparticle vaccine. Vaccine. 2020 Nov 3;38(47):7409-7413. [CrossRef]
- Ma Y, Fenton OS. An Efficacy and Mechanism Driven Study on the Impact of Hypoxia on Lipid Nanoparticle Mediated mRNA Delivery. J Am Chem Soc. 2023 May 24;145(20):11375-11386. [CrossRef]
- Reinhart AG, Osterwald A, Ringler P, et al. Investigations into mRNA Lipid Nanoparticles Shelf-Life Stability under Nonfrozen Conditions. Mol Pharm. 2023 Dec 4;20(12):6492-6503. [CrossRef]
- Szebeni J, Storm G, Ljubimova JY, et al. Applying lessons learned from nanomedicines to understand rare hypersensitivity reactions to mRNA-based SARS-CoV-2 vaccines. Nat Nanotechnol. 2022 Apr;17(4):337-346. [CrossRef]
- Hajiaghapour Asr M, Dayani F, Saedi Segherloo F, et al. Lipid Nanoparticles as Promising Carriers for mRNA Vaccines for Viral Lung Infections. Pharmaceutics. 2023 Apr 3;15(4):1127. [CrossRef]
- Rohde CM, Lindemann C, Giovanelli M, et al. Toxicological Assessments of a Pandemic COVID-19 Vaccine-Demonstrating the Suitability of a Platform Approach for mRNA Vaccines. Vaccines (Basel). 2023 Feb 11;11(2):417. [CrossRef]
- Zhu W, Wei L, Dong C, et al. cGAMP-adjuvanted multivalent influenza mRNA vaccines induce broadly protective immunity through cutaneous vaccination in mice. Mol Ther Nucleic Acids. 2022 Nov 9;30:421-437. [CrossRef]
- Austin LA, Smith JS, Nahas DD, et al. Split-Dose Administration Enhances Immune Responses Elicited by a mRNA/Lipid Nanoparticle Vaccine Expressing Respiratory Syncytial Virus F Protein. Mol Pharm. 2023 Jan 2;20(1):279-289. [CrossRef]
- Thaller A, Schmauder L, Frieß W, et al. SV-AUC as a stability-indicating method for the characterization of mRNA-LNPs. Eur J Pharm Biopharm. 2023 Jan;182:152-156. [CrossRef]
- Mu Z, Wiehe K, Saunders KO, et al. Ability of nucleoside-modified mRNA to encode HIV-1 envelope trimer nanoparticles. bioRxiv [Preprint]. 2021 Aug 9:2021.08.09.455714.
- Szebeni J, Kiss B, Bozó T, et al. Insights into the Structure of Comirnaty Covid-19 Vaccine: A Theory on Soft, Partially Bilayer-Covered Nanoparticles with Hydrogen Bond-Stabilized mRNA-Lipid Complexes. ACS Nano. 2023 Jul 25;17(14):13147-13157. [CrossRef]
- Messerian KO, Zverev A, Kramarczyk JF, et al. Pressure-dependent fouling behavior during sterile filtration of mRNA-containing lipid nanoparticles. Biotechnol Bioeng. 2022 Nov;119(11):3221-3229. [CrossRef]
- Shepherd SJ, Han X, Mukalel AJ, et al. Throughput-scalable manufacturing of SARS-CoV-2 mRNA lipid nanoparticle vaccines. Proc Natl Acad Sci U S A. 2023 Aug 15;120(33):e2303567120. [CrossRef]
- Lazaros G, Klein AL, Hatziantoniou S, et al. The Novel Platform of mRNA COVID-19 Vaccines and Myocarditis: Clues into the Potential Underlying Mechanism. Vaccine. 2021 Aug 16;39(35):4925-4927. [CrossRef]
- Han X, Alameh MG, Butowska K, et al. Adjuvant lipidoid-substituted lipid nanoparticles augment the immunogenicity of SARS-CoV-2 mRNA vaccines. Nat Nanotechnol. 2023 Sep;18(9):1105-1114. [CrossRef]
- Wang MM, Wappelhorst CN, Jensen EL, et al. Elucidation of lipid nanoparticle surface structure in mRNA vaccines. Sci Rep. 2023 Oct 5;13(1):16744. [CrossRef]
- Saunders KO, Pardi N, Parks R, et al. Lipid nanoparticle encapsulated nucleoside-modified mRNA vaccines elicit polyfunctional HIV-1 antibodies comparable to proteins in nonhuman primates. bioRxiv [Preprint]. 2020 Dec 31:2020.12.30.424745.
- Knudson CJ, Alves-Peixoto P, Muramatsu H, et al. Lipid-nanoparticle-encapsulated mRNA vaccines induce protective memory CD8 T cells against a lethal viral infection. Mol Ther. 2021 Sep 1;29(9):2769-2781. [CrossRef]
- Mu Z, Wiehe K, Saunders KO, et al. mRNA-encoded HIV-1 Env trimer ferritin nanoparticles induce monoclonal antibodies that neutralize heterologous HIV-1 isolates in mice. Cell Rep. 2022 Mar 15;38(11):110514. [CrossRef]
- Li Z, Zhang XQ, Ho W, et al. Enzyme-Catalyzed One-Step Synthesis of Ionizable Cationic Lipids for Lipid Nanoparticle-Based mRNA COVID-19 Vaccines. ACS Nano. 2022 Nov 22;16(11):18936-18950. [CrossRef]
- Saunders KO, Pardi N, Parks R, et al. Lipid nanoparticle encapsulated nucleoside-modified mRNA vaccines elicit polyfunctional HIV-1 antibodies comparable to proteins in nonhuman primates. NPJ Vaccines. 2021 Apr 9;6(1):50. [CrossRef]
- Willis E, Pardi N, Parkhouse K, et al. Nucleoside-modified mRNA vaccination partially overcomes maternal antibody inhibition of de novo immune responses in mice. Sci Transl Med. 2020 Jan 8;12(525):eaav5701. [CrossRef]
- Melzi E, Willis JR, Ma KM, et al. Membrane-bound mRNA immunogens lower the threshold to activate HIV Env V2 apex-directed broadly neutralizing B cell precursors in humanized mice. Immunity. 2022 Nov 8;55(11):2168-2186.e6. [CrossRef]
- Ma Y, Fenton OS. A Unified Strategy to Improve Lipid Nanoparticle Mediated mRNA Delivery Using Adenosine Triphosphate. J Am Chem Soc. 2023 Sep 13;145(36):19800-19811. [CrossRef]
- Everton E, Rizvi F, Smith AR, et al. Transient yet Robust Expression of Proteins in the Mouse Liver via Intravenous Injection of Lipid Nanoparticle-encapsulated Nucleoside-modified mRNA. Bio Protoc. 2021 Oct 5;11(19):e4184. [CrossRef]
- Lindgren G, Ols S, Liang F, et al. Induction of Robust B Cell Responses after Influenza mRNA Vaccination Is Accompanied by Circulating Hemagglutinin-Specific ICOS+ PD-1+ CXCR3+ T Follicular Helper Cells. Front Immunol. 2017 Nov 13;8:1539. [CrossRef]
- Shirane D, Tanaka H, Sakurai Y, et al. Development of an Alcohol Dilution-Lyophilization Method for the Preparation of mRNA-LNPs with Improved Storage Stability. Pharmaceutics. 2023 Jun 26;15(7):1819. [CrossRef]
- Kiaie SH, Majidi Zolbanin N, Ahmadi A, et al. Recent advances in mRNA-LNP therapeutics: immunological and pharmacological aspects. J Nanobiotechnology. 2022 Jun 14;20(1):276.
- Ndeupen S, Qin Z, Jacobsen S, et al. The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory. iScience. 2021 Dec 17;24(12):103479. [CrossRef]
- Zong Y, Lin Y, Wei T, et al. Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy. Adv Mater. 2023 May 17:e2303261. [CrossRef]
- Ramos da Silva J, Bitencourt Rodrigues K, Formoso Pelegrin G, et al. Single immunizations of self-amplifying or non-replicating mRNA-LNP vaccines control HPV-associated tumors in mice. Sci Transl Med. 2023 Mar 8;15(686):eabn3464. [CrossRef]
- Ogawa K, Kato N, Yoshida M, et al. Focused ultrasound/microbubbles-assisted BBB opening enhances LNP-mediated mRNA delivery to brain. J Control Release. 2022 Aug;348:34-41. [CrossRef]
- Wollner CJ, Richner M, Hassert MA, et al. A Dengue Virus Serotype 1 mRNA-LNP Vaccine Elicits Protective Immune Responses. J Virol.2021 May 24;95(12):e02482-20. [CrossRef]
- Ripoll M, Bernard MC, Vaure C, et al. An imidazole modified lipid confers enhanced mRNA-LNP stability and strong immunization properties in mice and non-human primates. Biomaterials. 2022 Jul;286:121570. [CrossRef]
- Kon E, Levy Y, Elia U, et al. A single-dose F1-based mRNA-LNP vaccine provides protection against the lethal plague bacterium. Sci Adv. 2023 Mar 10;9(10):eadg1036. [CrossRef]
- Granados-Riveron JT, Aquino-Jarquin G. Engineering of the current nucleoside-modified mRNA-LNP vaccines against SARS-CoV-2. Biomed Pharmacother. 2021 Oct;142:111953. [CrossRef]
- Qin Z, Bouteau A, Herbst C, et al. Pre-exposure to mRNA-LNP inhibits adaptive immune responses and alters innate immune fitness in an inheritable fashion. bioRxiv [Preprint]. 2022 Aug 20:2022.03.16.484616.
- Qin Z, Bouteau A, Herbst C, et al. Pre-exposure to mRNA-LNP inhibits adaptive immune responses and alters innate immune fitness in an inheritable fashion. PLoS Pathog. 2022 Sep 2;18(9):e1010830. [CrossRef]
- Baldeon Vaca G, Meyer M, Cadete A, et al. Intranasal mRNA-LNP vaccination protects hamsters from SARS-CoV-2 infection. Sci Adv. 2023 Sep 22;9(38):eadh1655. [CrossRef]
- Hayashi CTH, Cao Y, Clark LC, et al. mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines. 2022 Dec 1;7(1):155. [CrossRef]
- Qiu K, Duan X, Mao M, et al. mRNA-LNP vaccination-based immunotherapy augments CD8+ T cell responses against HPV-positive oropharyngeal cancer. NPJ Vaccines. 2023 Sep 29;8(1):144. [CrossRef]
- Valentin A, Bergamaschi C, Rosati M, et al. Comparative immunogenicity of an mRNA/LNP and a DNA vaccine targeting HIV gag conserved elements in macaques. Front Immunol. 2022 Jul 22;13:945706. [CrossRef]
- Aldrich C, Leroux-Roels I, Huang KB, et al. Proof-of-concept of a low-dose unmodified mRNA-based rabies vaccine formulated with lipid nanoparticles in human volunteers: A phase 1 trial. Vaccine. 2021 Feb 22;39(8):1310-1318. [CrossRef]
- Daly O, Mahiny AJ, Majeski S, et al. ASL mRNA-LNP Therapeutic for the Treatment of Argininosuccinic Aciduria Enables Survival Benefit in a Mouse Model. Biomedicines. 2023 Jun 16;11(6):1735. [CrossRef]
- Zhang M, Sun J, Li M, et al. Modified mRNA-LNP Vaccines Confer Protection against Experimental DENV-2 Infection in Mice. Mol Ther Methods Clin Dev. 2020 Jul 21;18:702-712. [CrossRef]
- Zhang R, Shao S, Piao Y, et al. Esterase-Labile Quaternium Lipidoid Enabling Improved mRNA-LNP Stability and Spleen-Selective mRNA Transfection. Adv Mater. 2023 Nov;35(46):e2303614. [CrossRef]
- Naderi Sohi A, Kiani J, Arefian E, et al. Development of an mRNA-LNP Vaccine against SARS-CoV-2: Evaluation of Immune Response in Mouse and Rhesus Macaque. Vaccines (Basel). 2021 Sep 10;9(9):1007. [CrossRef]
- Guéguen C, Ben Chimol T, Briand M, et al. Evaluating how cationic lipid affects mRNA-LNP physical properties and biodistribution. Eur J Pharm Biopharm. 2023 Aug 12:S0939-6411(23)00205-9. [CrossRef]
- He L, Sun W, Yang L, et al. A multiple-target mRNA-LNP vaccine induces protective immunity against experimental multi-serotype DENV in mice. Virol Sin. 2022 Oct;37(5):746-757. [CrossRef]
- Hsu FF, Liang KH, Kumari M, et al. An efficient approach for SARS-CoV-2 monoclonal antibody production via modified mRNA-LNP immunization. Int J Pharm. 2022 Nov 5;627:122256. [CrossRef]
- Zhang Y, Li D, Shen Y, et al. Immunization with a novel mRNA vaccine, TGGT1_216200 mRNA-LNP, prolongs survival time in BALB/c mice against acute toxoplasmosis. Front Immunol. 2023 Apr 14;14:1161507. [CrossRef]
- Zhang L, More KR, Ojha A, et al. Effect of mRNA-LNP components of two globally-marketed COVID-19 vaccines on efficacy and stability. NPJ Vaccines. 2023 Oct 11;8(1):156. [CrossRef]
- Chen T, Zhu S, Wei N, et al. Protective Immune Responses Induced by an mRNA-LNP Vaccine Encoding prM-E Proteins against Japanese Encephalitis Virus Infection. Viruses. 2022 May 24;14(6):1121. [CrossRef]
- Mao S, Li S, Zhang Y, et al. A highly efficient needle-free-injection delivery system for mRNA-LNP vaccination against SARS-CoV-2. Nano Today. 2023 Feb;48:101730. [CrossRef]
- Ndeupen S, Qin Z, Igyártó BZ. Single-cell suspension preparation from murine organs following in vivo mRNA-LNP exposure. STAR Protoc. 2022 May 18;3(2):101350. [CrossRef]
- Golubovskaya V, Sienkiewicz J, Sun J, et al. mRNA-Lipid Nanoparticle (LNP) Delivery of Humanized EpCAM-CD3 Bispecific Antibody Significantly Blocks Colorectal Cancer Tumor Growth. Cancers (Basel). 2023 May 22;15(10):2860. [CrossRef]
- Patel N, Davis Z, Hofmann C, et al. Development and Characterization of an In Vitro Cell-Based Assay to Predict Potency of mRNA-LNP-Based Vaccines. Vaccines (Basel). 2023 Jul 10;11(7):1224. [CrossRef]
- Rizvi F, Lee YR, Diaz-Aragon R, et al. VEGFA mRNA-LNP promotes biliary epithelial cell-to-hepatocyte conversion in acute and chronic liver diseases and reverses steatosis and fibrosis. Cell Stem Cell. 2023 Nov 20:S1934-5909(23)00392-2. [CrossRef]
- Moyles IR, Korosec CS, Heffernan JM. Determination of significant immunological timescales from mRNA-LNP-based vaccines in humans. J Math Biol. 2023 Apr 30;86(5):86. [CrossRef]
- Lamoot A, Lammens J, De Lombaerde E, et al. Successful batch and continuous lyophilization of mRNA LNP formulations depend on cryoprotectants and ionizable lipids. Biomater Sci. 2023 Jun 13;11(12):4327-4334. [CrossRef]
- Li D, Zhang Y, Li S, et al. A novel Toxoplasma gondii TGGT1_316290 mRNA-LNP vaccine elicits protective immune response against toxoplasmosis in mice. Front Microbiol. 2023 Mar 21;14:1145114. [CrossRef]
- Sáez-Llorens X, Lanata C, Aranguren E, et al. Safety and immunogenicity of mRNA-LNP COVID-19 vaccine CVnCoV in Latin American adults: A phase 2 randomized study. Vaccine X. 2022 Aug;11:100189. [CrossRef]
- Carter B, Huang P, Liu G, et al. A pan-variant mRNA-LNP T cell vaccine protects HLA transgenic mice from mortality after infection with SARS-CoV-2 Beta. Front Immunol. 2023 Mar 9;14:1135815. [CrossRef]
- Ndeupen S, Bouteau A, Herbst C, et al. Langerhans cells and cDC1s play redundant roles in mRNA-LNP induced protective anti-influenza and anti-SARS-CoV-2 responses. bioRxiv [Preprint]. 2021 Aug 2:2021.08.01.454662.
- Ndeupen S, Bouteau A, Herbst C, et al. Langerhans cells and cDC1s play redundant roles in mRNA-LNP induced protective anti-influenza and anti-SARS-CoV-2 immune responses. PLoS Pathog. 2022 Jan 24;18(1):e1010255. [CrossRef]
- Hayashi CTH, Cao Y, Clark LC, et al. Author Correction: mRNA-LNP expressing PfCSP and Pfs25 vaccine candidates targeting infection and transmission of Plasmodium falciparum. NPJ Vaccines. 2023 Aug 11;8(1):115. [CrossRef]
- Bavli Y, Chen BM, Gross G, Anti-PEG antibodies before and after a first doseet al. of Comirnaty (mRNA-LNP-based SARS-CoV-2 vaccine). J Control Release. 2023 Feb;354:316-322. [CrossRef]
- Hermosilla J, Alonso-García A, Salmerón-García A, et al. Analysing the In-Use Stability of mRNA-LNP COVID-19 Vaccines Comirnaty™ (Pfizer) and Spikevax™ (Moderna): A Comparative Study of the Particulate. Vaccines (Basel). 2023 Oct 25;11(11):1635. [CrossRef]
- Egan KP, Awasthi S, Tebaldi G, et al. A Trivalent HSV-2 gC2, gD2, gE2 Nucleoside-Modified mRNA-LNP Vaccine Provides Outstanding Protection in Mice against Genital and Non-Genital HSV-1 Infection, Comparable to the Same Antigens Derived from HSV-1. Viruses. 2023 Jun 30;15(7):1483. [CrossRef]
- Zhang Y, Li S, Chu H, et al. A novel mRNA vaccine, TGGT1_278620 mRNA-LNP, prolongs the survival time in BALB/c mice with acute toxoplasmosis. Microbiol Spectr. 2023 Dec 1:e0286623. [CrossRef]
- Sáez-Llorens X, Lanata C, Aranguren E, et al. Corrigendum to "Safety and immunogenicity of mRNA-LNP COVID-19 vaccine CVnCoV in Latin American adults: A phase 2 randomized study" [Vaccine: X 11 (2022) 100189]. Vaccine X. 2023 Aug;14:100307. [CrossRef]
- Vlatkovic I. Non-Immunotherapy Application of LNP-mRNA: Maximizing Efficacy and Safety. Biomedicines. 2021 May 10;9(5):530. [CrossRef]
- Maugeri M, Nawaz M, Papadimitriou A, et al. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Nat Commun. 2019 Sep 24;10(1):4333. [CrossRef]
- Kenjo E, Hozumi H, Makita Y, et al. Low immunogenicity of LNP allows repeated administrations of CRISPR-Cas9 mRNA into skeletal muscle in mice. Nat Commun. 2021 Dec 8;12(1):7101. [CrossRef]
- Álvarez-Benedicto E, Farbiak L, Márquez Ramírez M, et al. Optimization of phospholipid chemistry for improved lipid nanoparticle (LNP) delivery of messenger RNA (mRNA). Biomater Sci. 2022 Jan 18;10(2):549-559. [CrossRef]
- Peng L, Fang Z, Renauer PA, et al. Multiplexed LNP-mRNA vaccination against pathogenic coronavirus species. Cell Rep. 2022 Aug 2;40(5):111160. [CrossRef]
- Tsiambas E, Chrysovergis A, Papanikolaou V, et al. Impact of Ribosome Activity on SARS-CoV-2 LNP - Based mRNA Vaccines. Front Mol Biosci. 2021 Apr 20;8:654866. [CrossRef]
- Bahl K, Senn JJ, Yuzhakov O, et al. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol Ther. 2017 Jun 7;25(6):1316-1327. [CrossRef]
- Patel SK, Billingsley MM, Frazee C, et al. Hydroxycholesterol substitution in ionizable lipid nanoparticles for mRNA delivery to T cells. J Control Release. 2022 Jul;347:521-532. [CrossRef]
- Gao K, Li J, Song H, et al. In utero delivery of mRNA to the heart, diaphragm and muscle with lipid nanoparticles. Bioact Mater. 2023 Feb 17;25:387-398. [CrossRef]
- Ball RL, Hajj KA, Vizelman J, et al. Lipid Nanoparticle Formulations for Enhanced Co-delivery of siRNA and mRNA. Nano Lett. 2018 Jun 13;18(6):3814-3822. [CrossRef]
- Bogaert B, Sauvage F, Guagliardo R, et al. A lipid nanoparticle platform for mRNA delivery through repurposing of cationic amphiphilic drugs. J Control Release. 2022 Oct;350:256-270. [CrossRef]
- Huayamares SG, Lokugamage MP, Rab R, et al. High-throughput screens identify a lipid nanoparticle that preferentially delivers mRNA to human tumors in vivo. J Control Release. 2023 May;357:394-403. [CrossRef]
- Da Silva Sanchez AJ, Zhao K, Huayamares SG, et al. Substituting racemic ionizable lipids with stereopure ionizable lipids can increase mRNA delivery. J Control Release. 2023 Jan;353:270-277. [CrossRef]
- Swingle KL, Safford HC, Geisler HC, et al. Ionizable Lipid Nanoparticles for In Vivo mRNA Delivery to the Placenta during Pregnancy. J Am Chem Soc. 2023 Mar 1;145(8):4691-4706. [CrossRef]
- Attarwala H, Lumley M, Liang M, et al. Translational Pharmacokinetic/Pharmacodynamic Model for mRNA-3927, an Investigational Therapeutic for the Treatment of Propionic Acidemia. Nucleic Acid Ther. 2023 Apr;33(2):141-147. [CrossRef]
- Miao L, Lin J, Huang Y, et al. Synergistic lipid compositions for albumin receptor mediated delivery of mRNA to the liver. Nat Commun. 2020 May 15;11(1):2424. [CrossRef]
- Liang F, Lindgren G, Lin A, et al. Efficient Targeting and Activation of Antigen-Presenting Cells In Vivo after Modified mRNA Vaccine Administration in Rhesus Macaques. Mol Ther. 2017 Dec 6;25(12):2635-2647. [CrossRef]
- Ryals RC, Patel S, Acosta C, et al. The effects of PEGylation on LNP based mRNA delivery to the eye. PLoS One. 2020 Oct 29;15(10):e0241006. [CrossRef]
- Zhuang X, Qi Y, Wang M, et al. mRNA Vaccines Encoding the HA Protein of Influenza A H1N1 Virus Delivered by Cationic Lipid Nanoparticles Induce Protective Immune Responses in Mice. Vaccines (Basel). 2020 Mar 10;8(1):123. [CrossRef]
- Sinegra AJ, Evangelopoulos M, Park J, et al. Lipid Nanoparticle Spherical Nucleic Acids for Intracellular DNA and RNA Delivery. Nano Lett. 2021 Aug 11;21(15):6584-6591. [CrossRef]
- Zhang Y, Yan J, Hou X, et al. STING Agonist-Derived LNP-mRNA Vaccine Enhances Protective Immunity Against SARS-CoV-2. Nano Lett. 2023 Apr 12;23(7):2593-2600. [CrossRef]
- Dobrowolski C, Paunovska K, Schrader Echeverri E, et al. Nanoparticle single-cell multiomic readouts reveal that cell heterogeneity influences lipid nanoparticle-mediated messenger RNA delivery. Nat Nanotechnol. 2022 Aug;17(8):871-879. [CrossRef]
- Ickenstein LM, Garidel P. Lipid-based nanoparticle formulations for small molecules and RNA drugs. Expert Opin Drug Deliv. 2019 Nov;16(11):1205-1226. [CrossRef]
- Semple SC, Leone R, Barbosa CJ, et al. Lipid Nanoparticle Delivery Systems to Enable mRNA-Based Therapeutics. Pharmaceutics. 2022 Feb 11;14(2):398. [CrossRef]
- Tanaka H, Hagiwara S, Shirane D, et al. Ready-to-Use-Type Lyophilized Lipid Nanoparticle Formulation for the Postencapsulation of Messenger RNA. ACS Nano. 2023 Feb 14;17(3):2588-2601. [CrossRef]
- Tam A, Kulkarni J, An K, et al. Lipid nanoparticle formulations for optimal RNA-based topical delivery to murine airways. Eur J Pharm Sci. 2022 Sep 1;176:106234. [CrossRef]
- Hamilton AG, Swingle KL, Joseph RA, et al. Ionizable Lipid Nanoparticles with Integrated Immune Checkpoint Inhibition for mRNA CAR T Cell Engineering. Adv Healthc Mater. 2023 Dec;12(30):e2301515. [CrossRef]
- Shi D, Toyonaga S, Anderson DG. In Vivo RNA Delivery to Hematopoietic Stem and Progenitor Cells via Targeted Lipid Nanoparticles. Nano Lett. 2023 Apr 12;23(7):2938-2944. [CrossRef]
- Fang Z, Peng L, Filler R, et al. Omicron-specific mRNA vaccination alone and as a heterologous booster against SARS-CoV-2. bioRxiv [Preprint]. 2022 Feb 28:2022.02.14.480449.
- Radloff K, Gutbier B, Dunne CM, et al. Cationic LNP-formulated mRNA expressing Tie2-agonist in the lung endothelium prevents pulmonary vascular leakage. Mol Ther Nucleic Acids. 2023 Oct 29;34:102068. [CrossRef]
- August A, Brito L, Paris R, et al. Clinical Development of mRNA Vaccines: Challenges and Opportunities. Curr Top Microbiol Immunol. 2022;440:167-186. [CrossRef]
- Fedorowski JJ. Could amantadine interfere with COVID-19 vaccines based on the LNP-mRNA platform? Arch Med Sci. 2021 Mar 28;17(3):827-828. [CrossRef]
- Somiya M, Mine S, Yasukawa K, et al. Sex differences in the incidence of anaphylaxis to LNP-mRNA COVID-19 vaccines. Vaccine. 2021 Jun 8;39(25):3313-3314. [CrossRef]
- Wang W, Feng S, Ye Z, et al. Prediction of lipid nanoparticles for mRNA vaccines by the machine learning algorithm. Acta Pharm Sin B. 2022 Jun;12(6):2950-2962. [CrossRef]
- Leung J, Strong C, Badior KE, et al. Genetically engineered transfusable platelets using mRNA lipid nanoparticles. Sci Adv. 2023 Dec;9(48):eadi0508. [CrossRef]
- Novakowski S, Jiang K, Prakash G, et al. Delivery of mRNA to platelets using lipid nanoparticles. Sci Rep. 2019 Jan 24;9(1):552. [CrossRef]
- Sayers EJ, Peel SE, Schantz A, et al. Endocytic Profiling of Cancer Cell Models Reveals Critical Factors Influencing LNP-Mediated mRNA Delivery and Protein Expression. Mol Ther. 2019 Nov 6;27(11):1950-1962. [CrossRef]
- Wu L, Wang W, Tian J, et al. Engineered mRNA-expressed bispecific antibody prevent intestinal cancer via lipid nanoparticle delivery. Bioengineered. 2021 Dec;12(2):12383-12393. [CrossRef]
- Zeng Y, Escalona-Rayo O, Knol R, et al. Lipid nanoparticle-based mRNA candidates elicit potent T cell responses. Biomater Sci. 2023 Jan 31;11(3):964-974. [CrossRef]
- Wang Y, Si X, Feng Y, et al. Ionizable Lipids with Triazole Moiety from Click Reaction for LNP-Based mRNA Delivery. Molecules. 2023 May 12;28(10):4046. [CrossRef]
- Yeh TF, Lin C, Sung HC. A review of technological developments in lipid nanoparticle application for mRNA vaccination. Hum Vaccin Immunother. 2023 Aug 1;19(2):2256040. [CrossRef]
- Provine NM, Klenerman P. Adenovirus vector and mRNA vaccines: Mechanisms regulating their immunogenicity. Eur J Immunol. 2023 Jun;53(6):e2250022. [CrossRef]
- Qin J, Xue L, Gong N, et al. RGD peptide-based lipids for targeted mRNA delivery and gene editing applications. RSC Adv. 2022 Sep 7;12(39):25397-25404. [CrossRef]
- Goswami R, Chatzikleanthous D, Lou G, et al. Mannosylation of LNP Results in Improved Potency for Self-Amplifying RNA (SAM) Vaccines. ACS Infect Dis. 2019 Sep 13;5(9):1546-1558. [CrossRef]
- Vigil TN, Zhang-Hulsey D, Santos JL, et al. Expediting in vitro characterization of mRNA-based gene therapies via high-content fluorescent imaging. Anal Biochem. 2021 Aug 15;627:114259. [CrossRef]
- Zhang Y, Wang J, Xing H, et al. Enhanced immunogenicity induced by mRNA vaccines with various lipid nanoparticles as carriers for SARS-CoV-2 infection. J Mater Chem B. 2023 Aug 9;11(31):7454-7465. [CrossRef]
- Pine M, Arora G, Hart TM, et al. Development of an mRNA-lipid nanoparticle vaccine against Lyme disease. Mol Ther. 2023 Sep 6;31(9):2702-2714. [CrossRef]
- Huo H, Cheng X, Xu J, et al. A fluorinated ionizable lipid improves the mRNA delivery efficiency of lipid nanoparticles. J Mater Chem B. 2023 May 17;11(19):4171-4180. [CrossRef]
- Diaz-Trelles R, Perez-Garcia CG. Present and future of lipid nanoparticle-mRNA technology in phenylketonuria disease treatment. Int Rev Cell Mol Biol. 2022;372:159-174. [CrossRef]
- Long J, Yu C, Zhang H, et al. Novel Ionizable Lipid Nanoparticles for SARS-CoV-2 Omicron mRNA Delivery. Adv Healthc Mater. 2023 May;12(13):e2202590. [CrossRef]
- Chang DF, Court KA, Holgate R, et al. Telomerase mRNA Enhances Human Skin Engraftment for Wound Healing. Adv Healthc Mater. 2023 Aug 24:e2302029. [CrossRef]
- VanBlargan LA, Himansu S, Foreman BM, et al. An mRNA Vaccine Protects Mice against Multiple Tick-Transmitted Flavivirus Infections. Cell Rep. 2018 Dec 18;25(12):3382-3392.e3. [CrossRef]
- 탈ak MM, Kaur K, Yoo J, et al. Modified mRNA Formulation and Stability for Cardiac and Skeletal Muscle Delivery. Pharmaceutics. 2023 Aug 22;15(9):2176. [CrossRef]
- Cao W, Xia T. mRNA lipid nanoparticles induce immune tolerance to treat human diseases. Med Rev (Berl). 2023 Apr 14;3(2):180-183. [CrossRef]
- Bähr-Mahmud H, Ellinghaus U, Stadler CR, et al. Preclinical characterization of an mRNA-encoded anti-Claudin 18.2 antibody. Oncoimmunology. 2023 Oct 16;12(1):2255041. [CrossRef]
- Swingle KL, Billingsley MM, Bose SK, et al. Amniotic fluid stabilized lipid nanoparticles for in utero intra-amniotic mRNA delivery. J Control Release. 2022 Jan;341:616-633. [CrossRef]
- Yihunie W, Nibret G, Aschale Y. Recent Advances in Messenger Ribonucleic Acid (mRNA) Vaccines and Their Delivery Systems: A Review. Clin Pharmacol. 2023 Aug 3;15:77-98. [CrossRef]
- Szőke D, Kovács G, Kemecsei É, et al. Nucleoside-modified VEGFC mRNA induces organ-specific lymphatic growth and reverses experimental lymphedema. Nat Commun. 2021 Jun 8;12(1):3460. [CrossRef]
- Pardi N, Weissman D. Nucleoside Modified mRNA Vaccines for Infectious Diseases. Methods Mol Biol. 2017;1499:109-121. [CrossRef]
- Sang Y, Zhang Z, Liu F, et al. Monkeypox virus quadrivalent mRNA vaccine induces immune response and protects against vaccinia virus. Signal Transduct Target Ther. 2023 Apr 28;8(1):172. [CrossRef]
- Broudic K, Amberg A, Schaefer M, et al. Nonclinical safety evaluation of a novel ionizable lipid for mRNA delivery. Toxicol Appl Pharmacol. 2022 Sep 15;451:116143. [CrossRef]
- Fekete S, Doneanu C, Addepalli B, et al. Challenges and emerging trends in liquid chromatography-based analyses of mRNA pharmaceuticals. J Pharm Biomed Anal. 2023 Feb 5;224:115174. [CrossRef]
- Sun M, Dang UJ, Yuan Y, et al. Optimization of DOTAP/chol Cationic Lipid Nanoparticles for mRNA, pDNA, and Oligonucleotide Delivery. AAPS PharmSciTech. 2022 May 9;23(5):135. [CrossRef]
- McCrudden CM, Bennie L, Chambers P, et al. Peptide delivery of a multivalent mRNA SARS-CoV-2 vaccine. J Control Release. 2023 Oct;362:536-547. [CrossRef]
- Thran M, Mukherjee J, Pönisch M, et al. mRNA mediates passive vaccination against infectious agents, toxins, and tumors. EMBO Mol Med. 2017 Oct;9(10):1434-1447. [CrossRef]
- Zhang J, Shrivastava S, Cleveland RO, et al. Lipid-mRNA Nanoparticle Designed to Enhance Intracellular Delivery Mediated by Shock Waves. ACS Appl Mater Interfaces. 2019 Mar 20;11(11):10481-10491. [CrossRef]
- Nakamura T, Nakade T, Sato Y, et al. Delivering mRNA to a human NK cell line, NK-92 cells, by lipid nanoparticles. Int J Pharm. 2023 Apr 5;636:122810. [CrossRef]
- Huysmans H, Zhong Z, De Temmerman J, et al. Expression Kinetics and Innate Immune Response after Electroporation and LNP-Mediated Delivery of a Self-Amplifying mRNA in the Skin. Mol Ther Nucleic Acids. 2019 Sep 6;17:867-878. [CrossRef]
- Dong S, Wang J, Guo Z, et al. Efficient delivery of VEGFA mRNA for promoting wound healing via ionizable lipid nanoparticles. Bioorg Med Chem. 2023 Jan 15;78:117135. [CrossRef]
- 241. Zhang HL. Current status and patent prospective of lipid nanoparticle for mRNA delivery. Expert Opin Ther Pat. 2023 Feb;33(2):125-131. [CrossRef]
- Yamazaki K, Kubara K, Ishii S, et al. Lipid nanoparticle-targeted mRNA formulation as a treatment for ornithine-transcarbamylase deficiency model mice. Mol Ther Nucleic Acids. 2023 Jul 4;33:210-226. [CrossRef]
- Patel S, Ashwanikumar N, Robinson E, et al. Boosting Intracellular Delivery of Lipid Nanoparticle-Encapsulated mRNA. Nano Lett. 2017 Sep 13;17(9):5711-5718. [CrossRef]
- Olson KE, Namminga KL, Lu Y, et al. Granulocyte-macrophage colony-stimulating factor mRNA and Neuroprotective Immunity in Parkinson's disease. Biomaterials. 2021 May;272:120786. [CrossRef]
- Nawaz M, Heydarkhan-Hagvall S, Tangruksa B, et al. Lipid Nanoparticles Deliver the Therapeutic VEGFA mRNA In Vitro and In Vivo and Transform Extracellular Vesicles for Their Functional Extensions. Adv Sci (Weinh). 2023 Apr;10(12):e2206187. [CrossRef]
- Popowski KD, López de Juan Abad B, George A, et al. Inhalable exosomes outperform liposomes as mRNA and protein drug carriers to the lung. Extracell Vesicle. 2022 Dec;1:100002. [CrossRef]
- Safford HC, Swingle KL, Geisler HC, et al. Orthogonal Design of Experiments for Engineering of Lipid Nanoparticles for mRNA Delivery to the Placenta. Small. 2023 Aug 3:e2303568. [CrossRef]
- Lokugamage MP, Gan Z, Zurla C, et al. Mild Innate Immune Activation Overrides Efficient Nanoparticle-Mediated RNA Delivery. Adv Mater. 2020 Jan;32(1):e1904905. [CrossRef]
- Zhdanov VP. Kinetics of lipid-nanoparticle-mediated intracellular mRNA delivery and function. Phys Rev E. 2017 Oct;96(4-1):042406. [CrossRef]
- Hatit MZC, Dobrowolski CN, Lokugamage MP, et al. Nanoparticle stereochemistry-dependent endocytic processing improves in vivo mRNA delivery. Nat Chem. 2023 Apr;15(4):508-515. [CrossRef]
- Miao H, Huang K, Li Y, et al. Optimization of formulation and atomization of lipid nanoparticles for the inhalation of mRNA. Int J Pharm. 2023 Jun 10;640:123050. [CrossRef]
- Hunter MR, Cui L, Porebski BT, et al. Understanding Intracellular Biology to Improve mRNA Delivery by Lipid Nanoparticles. Small Methods. 2023 Sep;7(9):e2201695. [CrossRef]
- Yu X, Yu C, Wu X, et al. Validation of an HPLC-CAD Method for Determination of Lipid Content in LNP-Encapsulated COVID-19 mRNA Vaccines. Vaccines (Basel). 2023 May 4;11(5):937. [CrossRef]
- Aliakbarinodehi N, Gallud A, Mapar M, et al. Interaction Kinetics of Individual mRNA-Containing Lipid Nanoparticles with an Endosomal Membrane Mimic: Dependence on pH, Protein Corona Formation, and Lipoprotein Depletion. ACS Nano. 2022 Dec 27;16(12):20163-20173. [CrossRef]
- Xue L, Gong N, Shepherd SJ, et al. Rational Design of Bisphosphonate Lipid-like Materials for mRNA Delivery to the Bone Microenvironment. J Am Chem Soc. 2022 Jun 8;144(22):9926-9937. [CrossRef]
- van Rijn CJM, Vlaming KE, Bem RA, et al. Low energy nebulization preserves integrity of SARS-CoV-2 mRNA vaccines for respiratory delivery. Sci Rep. 2023 May 31;13(1):8851. [CrossRef]
- Gan Z, Lokugamage MP, Hatit MZC, et al. Nanoparticles containing constrained phospholipids deliver mRNA to liver immune cells in vivo without targeting ligands. Bioeng Transl Med. 2020 May 27;5(3):e10161. [CrossRef]
- Zha W, Wang J, Guo Z, et al. Efficient delivery of VEGF-A mRNA for promoting diabetic wound healing via ionizable lipid nanoparticles. Int J Pharm. 2023 Feb 5;632:122565. [CrossRef]
- Shepherd SJ, Warzecha CC, Yadavali S, et al. Scalable mRNA and siRNA Lipid Nanoparticle Production Using a Parallelized Microfluidic Device. Nano Lett. 2021 Jul 14;21(13):5671-5680. [CrossRef]
- Ye Z, Chen J, Zhao X, et al. In Vitro Engineering Chimeric Antigen Receptor Macrophages and T Cells by Lipid Nanoparticle-Mediated mRNA Delivery. ACS Biomater Sci Eng. 2022 Feb 14;8(2):722-733. [CrossRef]
- Bepperling A, Richter G. Determination of mRNA copy number in degradable lipid nanoparticles via density contrast analytical ultracentrifugation. Eur Biophys J. 2023 Jul;52(4-5):393-400. [CrossRef]
- Sarode A, Patel P, Vargas-Montoya N, et al. Inhalable dry powder product (DPP) of mRNA lipid nanoparticles (LNPs) for pulmonary delivery. Drug Deliv Transl Res. 2023 Aug 1. [CrossRef]
- Huang H, Zhang C, Yang S, et al. The investigation of mRNA vaccines formulated in liposomes administrated in multiple routes against SARS-CoV-2. J Control Release. 2021 Jul 10;335:449-456. [CrossRef]
- Elia U, Ramishetti S, Rosenfeld R, et al. Design of SARS-CoV-2 hFc-Conjugated Receptor-Binding Domain mRNA Vaccine Delivered via Lipid Nanoparticles. ACS Nano. 2021 Jun 22;15(6):9627-9637. [CrossRef]
- Yang D, Song CQ. The Delivery of ABE mRNA to the Adult Murine Liver by Lipid Nanoparticles (LNPs). Methods Mol Biol. 2023;2606:159-170. [CrossRef]
- Nakashima I, Saito S, Akahoshi E, et al. Non-viral inducible caspase 9 mRNA delivery using lipid nanoparticles against breast cancer: An in vitro study. Biochem Biophys Res Commun. 2022 Dec 20;635:144-153. [CrossRef]
- Wang T, Sung TC, Yu T, et al. Next-generation materials for RNA-lipid nanoparticles: lyophilization and targeted transfection. J Mater Chem B. 2023 Jun 14;11(23):5083-5093. [CrossRef]
- Takanashi A, Pouton CW, Al-Wassiti H. Delivery and Expression of mRNA in the Secondary Lymphoid Organs Drive Immune Responses to Lipid Nanoparticle-mRNA Vaccines after Intramuscular Injection. Mol Pharm. 2023 Aug 7;20(8):3876-3885. [CrossRef]
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).