Next Article in Journal
African Swine Fever Virus: A Review
Next Article in Special Issue
Selective RNA Labeling by RNA-Compatible Type II Restriction Endonuclease and RNA-Extending DNA Polymerase
Previous Article in Journal
The Impact of eHealth Interventions on the Improvement of Self-Care in Chronic Patients: An Overview of Systematic Reviews
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Delivery of mRNA Vaccines for Therapeutics

1
Fosun Pharma USA Inc., Boston, MA 02421, USA
2
Shanghai Fosun Pharmaceutical Industrial Development, Co., Ltd., Shanghai 200233, China
*
Author to whom correspondence should be addressed.
Life 2022, 12(8), 1254; https://doi.org/10.3390/life12081254
Submission received: 13 July 2022 / Revised: 5 August 2022 / Accepted: 15 August 2022 / Published: 17 August 2022
(This article belongs to the Special Issue Advances in Nucleic Acid Therapeutics)

Abstract

:
mRNA vaccines have been revolutionary in combating the COVID-19 pandemic in the past two years. They have also become a versatile tool for the prevention of infectious diseases and treatment of cancers. For effective vaccination, mRNA formulation, delivery method and composition of the mRNA carrier play an important role. mRNA vaccines can be delivered using lipid nanoparticles, polymers, peptides or naked mRNA. The vaccine efficacy is influenced by the appropriate delivery materials, formulation methods and selection of a proper administration route. In addition, co-delivery of several mRNAs could also be beneficial and enhance immunity against various variants of an infectious pathogen or several pathogens altogether. Here, we review the recent progress in the delivery methods, modes of delivery and patentable mRNA vaccine technologies.

1. Introduction

Messenger RNA (mRNA) is a transient intermediate between DNA and proteins, which was first discovered in 1961 [1]. Later, mRNA was in vitro transcribed and emerged as a candidate for therapeutic purposes, including immunotherapies, viral vaccines, genome editing and cellular reprogramming [1]. The desired therapeutic effects of mRNA can be achieved only if the mRNA molecule reaches a specific target cell and produces enough protein of interest [2]. The methods and delivery of mRNA intracellular delivery present a major barrier to the broader application of mRNA therapeutics [3]. The intracellular delivery of mRNA is more challenging because of the larger size of mRNA (300–1500 kDa) as compared to a smaller size (4–14 kDa) for small interference RNA (siRNA) and antisense oligonucleotide (ASO) [4,5]. The major barrier to mRNA delivery to the target cell is the cell membrane reaching the cytoplasm. The cell membrane is composed of a zwitterionic lipid bilayer and negatively charged phospholipids [6,7]. The negatively charged mRNA molecules face repulsion from the cell membrane, which is also negatively charged. mRNA is also susceptible to degradation by ribonucleases in the extracellular environment. Therefore, the mRNA molecule needs protection from degradation by nucleases and shields its negative charge [8,9].
A variety of mRNA delivery methods have been developed, such as direct injection of naked mRNA, lipid-based carriers, polymers and protein derivatives [10]. Compared to other delivery vehicles, lipid nanoparticles have been extensively studied for the delivery of small molecules, such as siRNA and mRNA [6].
The utility of lipid nanoparticles to deliver mRNA has been successfully demonstrated with COVID-19 vaccines, such as mRNA-1273 and BNT162b [11]. More than 10% of vaccines being tested for use against SARS-CoV2 in humans are mRNA-based vaccines, including mRNA-1273, BNT162b, CVnCoV, ARCoV, ARCT-02, ChulaCov19 and LNP-nCoV saRNA. Of all the seven mRNA vaccines in clinical trials, only mRNA-1273 and BNT162b have been approved for use in humans. Other LNP-mRNA formulations have also been used for genetic diseases, virus infections and cancer [12,13].
Despite the recent advances in mRNA vaccines, there is growing interest in developing improved safety and effective delivery methods for mRNA-based therapies. With the emergence of the COVID-19 global pandemic, lipid-nanoparticle-based mRNA vaccines have emerged as the fastest and most efficient tool for combating such infectious diseases [14]. mRNA vaccine delivery is important to achieve vaccine efficiency. In this review, we summarize the mRNA delivery strategy, including the delivery barrier, the routes of administration and delivery vehicles that have been utilized both in preclinical and clinical settings.

2. mRNA Therapeutic Delivery Barriers

mRNA-based vaccines have several benefits over other vaccines. mRNA vaccine sequences are precise and express a specific antigen leading to the induction of a directed immune response [15]. mRNA vaccines promote both humoral and cellular immune responses and induce the innate immune system. Additionally, nucleoside modification in the mRNA sequence reduces its inflammatory capacity. Therefore, mRNA-based vaccines are safe for delivery when compared to virus-based vaccines. The safety concerns for the viral contaminants are minimized since production is based on an in vitro cell-free transcription reaction [14].
In addition to the benefits of mRNA vaccines, there are several challenges associated with mRNA delivery that need to be addressed. The development of optimal delivery systems to protect mRNA and efficient delivery into the cells remains an area of active research [16]. mRNA vaccines might cause side effects, such as heart and renal failure, allergy and infarction [17]. mRNA vaccines may be degraded quickly in the body after administration or cause a cytokine storm, which poses a challenge to their delivery.
The delivery of mRNA into the cell is the most challenging part of mRNA therapeutics. The mRNA must cross several extracellular and intracellular barriers before it arrives in the cytoplasm or nucleus of its target cell [7,18].
The extracellular barriers include the cell membrane, which is composed of a lipid bilayer of a zwitterionic negatively charged phospholipid, ion pumps and channels, which maintain negative potential, thus, making it difficult for the mRNA to enter the cell [19]. Additional extracellular barriers include the presence of exonucleases or RNases in skin and blood. These exonucleases digest mRNA before its entry into the cell [7].
The intracellular barriers include endosomal escape, RNA sensors and endonucleases. Once the mRNA-based vaccine encounters the plasma membrane, it is engulfed and processed by an endocytic pathway to be released into the cell. However, a small percentage of LNPs evade the endocytic pathway due to the disruption of the endosomal membrane caused by the protonation of the LNP residual amines [10,20]. This leads to the premature release of LNP-mRNA cargo into the cell affecting the efficiency of the mRNA vaccine [21]. Another barrier to mRNA vaccines is the recognition of the mRNA by cytosolic innate sensors, such as toll-like receptors (TLRs), TLR 3 and TLR7. The TLRs bind to the mRNA and activate the inflammatory pathways, leading to the production of type I interferon and other inflammatory mediators, which results in inhibition of the cellular translation [22,23]. Additionally, intracellular RNases also act as a barrier by degradation of mRNA, even before it is translated to produce the antigen in the cell [24,25].

3. Routes of Administration

The route of mRNA vaccine delivery is essential to determine the efficacy of the vaccination. The anatomical and physiological properties of the site of vaccination, such as skin, lymphoid organ or muscle, affect the safety and efficacy of the vaccine [24,26]. The vaccines can be administered via either systemic or local applications [27,28,29].
Systemic delivery is the delivery route where the vaccine is directly injected into the bloodstream to reach and affect all cells in the body [18,30], such as intravenous injections [31]. Local injections are the mode of delivery where the vaccine is directly administered at the site of action. This route of delivery reduces the risk of side effects associated with systemic delivery (Figure 1). There is also much interest in targeted delivery [32]. This mode of delivery aims at direct injection into the target tissue or the organ. The intranodal injection is such an example of targeted delivery.

3.1. Intravenous Injections

Intravenous (IV) injections deliver mRNA vaccines into the systemic circulation in the body. IV injection provides the largest volume among all the other routes of vaccine administration. It was also observed that the total amount of antigen produced by IV is the highest when compared to the other routes of vaccine delivery [2,10]. IV injections allow for the direct access of mRNA vaccines to the immune cells and lymphoid organs in the circulatory system, which then leads to increased efficacy of the mRNA vaccines [33]. Even though IV is an efficacious method, there are several disadvantages, which include hindrance to vaccine delivery in the bloodstream by plasma proteins, enzymes and mechanical forces [34]. In addition, the mRNA and its delivery carriers might introduce systemic side effects, including spleen injury and depletion of the lymphocytes [18].

3.2. Subcutaneous Injections

Subcutaneous (SC)-injection-based mRNA vaccines are administered under the epidermis and dermis in the subcutis layer of the skin [35]. This layer of skin constitutes a loose network of adipose tissues and few immune cells. The loose adipose tissue at the injection site allows for a larger injection volume, which lowers the pressure and reduces pain at the site [36]. The larger injection volume also allows for efficient draining of the immune system. However, one of the disadvantages of the SC injection is that the rate of absorption is low and unintended degradation of the mRNA may occur [36,37].

3.3. Intramuscular Injections

Intramuscular (IM) injections are the most used route of administration for mRNA vaccines. Muscles are composed of a large network of blood vessels that recruit and circulate various immune cells, such as antigen-presenting cells (APCs), to the site of the injection [38]. IM-injection-based vaccines are administered into muscles with deeper tissue under the dermal and subcutaneous layer of the skin [39]. It has been shown that the IM-administered LNP-mRNA remains at the site of injection and in the draining lymph nodes for ~28 h [37,40]. IM has been used for mRNA vaccines against the RBD of SARS-CoV-2 virus and has been successful in conferring immunity against the virus [41].

3.4. Intradermal Injections

Intradermal injections are administered in the dermis layer of the skin, a dense connective tissue, which has vascular and lymphatic vessels, which help transport the mRNA vaccines and APCs to the draining lymph nodes to activate B and T cells [42].

3.5. Intranodal Injections

Intranodal injections deliver the mRNA vaccines in the peripheral lymphoid organs where APCs and immune cells, such as B and T cells, interact. The APCs in the lymphoid organs readily engulf the mRNA vaccine [43]. There are reports on the efficacy of intranodal-based DNA, peptide and protein vaccines but its efficacy for mRNA vaccines remains to be explored [44]. Additionally, the administration of the intranodal mRNA vaccines needs the guidance of ultrasound for the administration [45].
Other delivery methods include intranasal injection or inhalation-based delivery of mRNA vaccines, which are studied in respiratory delivery [46,47,48]. The inhaled materials encounter pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), which derive APCs to take up the antigen. The dendritic cells then migrate via lymphatic vessels to the lymph nodes where the antigen is presented via MHC II complexes to naïve B and T cells. In addition, the respiratory system has inducible bronchus-associated lymphoid tissue (iBALT), which consists of B-cell follicles, plasma cells, T cells and APCs. Antigens are then presented to both the effector and naïve B and T cells. These tertiary lymphoid tissues are interconnected with the mucosal-associated lymphoid tissue (MALT) [47,49].

4. Delivery Vehicle

4.1. Naked mRNA

Naked-mRNA-based vaccines are delivered by dissolving the mRNA into a buffer and then injecting the mRNA solution directly into the body (Figure 2A) [24]. Although naked mRNA cannot diffuse across the intracellular membrane, the mechanism for its internalization remains unclear [50]. Initially, it was proposed to be internalized by a cellular process, known as micropinocytosis [51,52]. Some studies have suggested that the internalization of naked mRNA is facilitated by mechanical forces, such as hydrostatic pressure. It is speculated that hydrostatic pressure may lead to the disruption of the cell membrane and facilitate cytosolic delivery of nucleic acids [53]. Some of the benefits of naked mRNA-based vaccines include storage stability and intrinsic immunogenicity [54]. Naked mRNA can be easily stored at 4 °C for up to 10 months in 10% trehalose upon freeze drying [54]. Similarly, the intrinsic immunogenicity is advantageous because it triggers RNA sensors, such as TLRs, RIG-I, PKR and IFIT1, which leads to the activation of NF KB, type I IFN pathways and the release of cytokines. Some reports have also indicated that naked mRNA induces an innate immune response [55]. Unmodified naked mRNA is considered a strong stimulator of TLR3/7/8 and PKR, although mRNA translation might be inactivated by certain RNA sensors in the cytosol [56,57]. In addition, naked mRNA vaccines are also susceptible to RNase degradation and intracellular delivery [58,59]. These obstacles can be removed by local administration of mRNA, such as intramuscular, intranodal, intratracheal, intradermal and intranasal routes, to minimize their contact with RNases in the bloodstream [43,56,60,61,62,63,64,65]. Naked mRNAs have been tested in clinical trials against diseases, such as melanoma, influenza and HIV-1 virus (Table 1) [66,67].

4.2. Electroporation for Dendritic-Cell-Based mRNA Vaccines

Antigen-presenting cells (APCs) are responsible for the internalization, processing and presentation of antigens to lymphocytes. Dendritic cells (DCs) are types of APCs that present processed antigens from microorganisms, tumor cells and virus-infected cells to T cells for the generation of the immune response [68,69,70]. DCs are suitable vaccination targets because of their migration to T cells in the lymph nodes, high expression of MHC, co-stimulators and cytokines [71]. Electroporation disrupts the cell membrane by generating electric shock for intracellular nucleic acid delivery. Delivery efficiency can be improved by adjusting voltage, capacitance, resistance and other factors, such as cell number, density, RNA quantity and pulse time [53,72,73]. Electroporation has been used for DC-based mRNA vaccines in clinical trials [74,75,76].

4.3. Peptide-Based Delivery Protamine

Peptides have been used as delivery carriers for mRNA vaccines [77,78]. To serve as delivery carriers, the peptides should contain strings of positively charged amino acids, such as lysine and arginine. This allows for the formation of electrostatic interactions between positively charged peptides and negatively charged mRNA, thus, enabling a spontaneous complex formation [79,80,81]. Protamines are advantageous as carriers of mRNA vaccines because they protect the mRNA and make it less susceptible to being degraded by RNases [82,83]. The protamine–mRNA complex has high adjuvant activity. The complex is immunogenic via activation because of its structural similarity to viral RNA genome [84,85]. The feasibility of the mRNA protamine complex was tested with β-galactosidase mRNA—protamine—which was injected into a glioblastoma tumor. It was observed that the mRNA complexed with protamine was poorly translated [86,87].

4.4. Polymer-Based Delivery

Polymers are functional materials that can deliver mRNA vaccines. Like protamines, polymers protect mRNA from RNase degradation [3]. Polymer-based mRNA nanoparticles have high polydispersity and to stabilize this, formulation modifications, such as incorporating lipid chains, hyperbranched groups and biodegradable units, are being explored. Cationic polymers include polyethyleneimine (PEI), polyamidoamine (PAMAM) dendrimer and polysaccharides [88,89,90,91]. PEI has been a widely used polymeric material for mRNA delivery and can be prepared by directly mixing PEI with RNA solution. PEI formulation was used to deliver a HIV gp120 mRNA-based intranasal vaccine in mice [92,93]. In addition, PEI-based formulation was used to deliver HIV-1 Gag and Pol antigens to produce T-cell response against HIV infections upon intramuscular vaccination in mice [93]. This vaccination approach protected mice against the viral challenge. PEI formulation has also been used for the delivery of self-amplifying mRNA encoding hemagglutinin antigens from various influenza strains for immunization in mice models [90]. Even though PEI has shown in vivo efficacy, the possible toxicity has hindered its development [3].
Another polymer that has been utilized is polyamidoamine (PAMAM), which is a cationic polymer. The PAMAM dendrimer has been used to deliver intramuscular self-amplifying mRNA-based vaccination against Toxoplasma gondii, Ebola and H1N1 influenza virus [91,94].
Like PAMAM, Chitosan, which is a polysaccharide material, has been used to condense and deliver self-amplifying mRNAs encoding hemagglutinin and nucleoprotein for influenza virus [8].
Polymer materials for the delivery of mRNA vaccines have been proven to be efficient in preclinical studies [95]. However, new functional polymers, with improved biodegradability and delivery efficacy, are desirable before the translation of polymer-based mRNA vaccines in the clinic.

4.5. Lipid-Based Delivery

Lipid Nanoparticles (LNPs)

Lipid-derived nanoparticles are widely used for in vivo delivery of mRNA vaccines [10,21,24,96]. They are composed of nano-sized particulates that are composed of synthetic lipid materials. LNP-based mRNA vaccines encapsulate RNA and protect it from RNAse-mediated degradation (Figure 2A) [21,97]. In addition, LNPs deliver mRNA molecules effectively through endocytosis mechanisms (Figure 2B). LNPs are generally composed of a functional lipid component that is crucial for the intracellular RNA delivery [98,99,100]. The cationic or ionizable lipid materials, such as 1,2-di-O-octadecyl-3-trimethylammonium propane (DOTMA), N, N-Dimethyl-2,3-bis[(9Z,12Z)-octadeca-9,12-dienyloxy]propan-1-amine (DLinDMA) and N1, N3, N5 -tris(3-(didodecylamino)propyl)benzene-1,3,5-tricarboxamide (TT3), with one or multiple amino groups, can be positively charged at a certain pH, which helps encapsulate the negatively charged mRNA molecules via electrostatic interactions and associate with the cell membrane [26,101,102]. Further, the ionizable cationic lipids interact with the anionic endosome membrane, which leads to the formation of a disruptive non-bilayer structure, which leads to the release of the mRNA into the cytosol. It has been indicated that the hydrophilic head of the lipid material determines the acid dissolution constant (pKa) and influences the delivery efficiency of the LNPs [103]. Although the lipid materials enhance efficacy, they might have some side effects on the cells [104]. The polyethylene glycol (PEG) lipid conjugates stabilize the nanoparticles during preparation and prolong the circulation time in vivo, which leads to adverse effects, such as anaphylaxis, that can lead to rashes, shortness of breath and plummeting blood pressure [105,106,107,108]. Additionally, the cyclic amino head groups of LNPs bind directly to the stimulator of interferon gene (STING) protein and activate the downstream signaling pathway, which leads to enhanced immune response [109]. The delivery routes of mRNA vaccines may impact their efficacy by affecting the distribution pattern and the expression pattern of the encapsulated mRNA [21]. The local injections, such as intramuscular, intradermal and intranasal administration, lead to infiltration of antigen-presenting cells, which stimulates strong and prolonged local expression [21,110]. LNPs were used to deliver the COVID-19 mRNA vaccine. The TT3-LNP was used to deliver the receptor-binding domain of SARS-CoV-2 intramuscularly, which led to the expression of the antigen in the muscle tissue [26]. Overall, LNP-based mRNA vaccines have efficacy in preventing infectious diseases and providing immunity [111].

4.6. Cationic Nanoemulsion

Cationic nanoemulsion (CNE) utilizes nanoemulsion with cationic lipids for RNA delivery. Nanoemulsion has hydrophobic and hydrophilic surfactants to stabilize the oil core in the aqueous phase, thereby generating particles. Nanoemulsions are produced using methods, such as vigorous agitation, ultrasound and microfluidics [112]. One of the oils in water nanoemulsions approved by the FDA is MF59, which was used as an adjuvant with inactivated flu vaccine for the elderly. MF59 consists of squalene, sorbitan trioleate, polyoxyethylene, sorbitan monooleate and citrate buffer [113]. The efficacy of vaccines by MF59 is enhanced by the MyD88-mediated release of chemokines and recruitment of immune cells [114,115,116]. CNEs have been used for the delivery of mRNA vaccines against bacterial and viral infections [117,118,119,120]. Three chimeric MF59-CNE-based mRNA vaccines against the respiratory syncytial virus (RSV), HIV and human cytomegalovirus (CMV) were intramuscularly injected into mice, rabbits and rhesus macaques [117]. The vaccines induced high antigen-specific IgG titer and an efficient leukocyte infiltration [117]. Overall, CNE is efficacious as a delivery method in preclinical studies; however, its efficacy in clinical trials remains to be evaluated.
Table 1. mRNA vaccines in various animal models.
Table 1. mRNA vaccines in various animal models.
Delivery Method Routes of Administration Target Immune Response Animal Model References
Naked mRNAIntramuscularTumorsHumoralMice[60]
IntramuscularInfluenza, RSV, EncephalitisHumoralMice[61]
IntradermalInfluenzaHumoral/CellularMice, Human[56]
IntranodalInfluenzaHumoralMice[43]
IntranasalTuberculosisHumoralMice[65]
DendrimerIntramuscularEbola, Influenza and ToxoplasmaHumoral/CellularMice[91]
IntramuscularZikaHumoral/CellularMice[121]
ProtamineIntradermal, IntramuscularRabies virusHumoralMice, Pigs, and human[82]
IntradermalInfluenzaHumoral/CellularMice, Ferrets, and pigs[122]
IntradermalProstate cancerCellularHuman[123]
PolymerIntramuscularInfluenzaHumoralMice[124]
IntranasalHIV-gp120HumoralMice
IntramuscularHIV-Gag and PolHumoral/CellularMice[93]
SubcutaneousZikaHumoral/CellularMice, Rabbit[90]
Lipid nanoparticleIntradermalZikaHumoralMice, Primates[12]
IntramuscularEbolaHumoralGuinea pigs[125]
IntradermalHIV-EnvHumoral/CellularMice[126]
IntravenousHIV-IgGHumoralMice[127]
IntramuscularSARS-CoV2HumoralHuman[128,129]
Cationic nanoemulsionSub cutaneousHIV—GagHumoralMice[130]
IntramuscularRSV, CMV, HIVHumoralMice, Rabbit, Macaques[117]
IntramuscularEncephalitisHumoralMice[120]
Virus-like replicon particleIntradermalDengueHumoralMacaques[131]
Intravenous, IntramuscularInfluenzaHumoral/CellularMice, Swine[132]
IntramuscularHIVHumoral/CellularMice[133]
Subcutaneous, IntramuscularEbolaCellularPrimates[134]
IntramuscularSARS-CoVHumoralMice[135]
IntranasalMERS-CoVHumoralMice[136]
IntradermalRSVHumoralPrimates[137]
Subcutaneous, intramuscularCMVHumoralHuman[138]

4.7. Virus-Like Replicon Particle (VRP)

Viral particles can be used to package and deliver antigen-encoding self-amplifying mRNA in cytoplasm like a virus [139]. Self-amplifying mRNA can then replicate and efficiently express the designated antigens. VRPs are efficient in cytoplasmic delivery of RNA payload by viral vectors [92,140]. This is because viruses internalize and release their genomes into cells via different pathways with high efficiency (Figure 2B) [141,142]. The most used VRPs for vaccines are single-stranded RNAs, including alphavirus, flavivirus, rhabdovirus and measles virus [139]. VRP vaccines were injected intradermally in non-human primates to produce immunity against the Venezuelan equine encephalitis virus (VEEV) [131]. Similarly, a Kunjin virus-derived VRP expressing GM-CSF was injected intratumorally in mice with colon carcinoma, which led to complete removal of the primary tumor and a reduction in lung metastases [143]. However, there are challenges for VRP-based mRNA vaccines, including the process of generating VRPs, which limits scaling up in the production of VRPs [144,145]. Another challenge is that, sometimes, there is antibody generation against viral vectors as well, which hinders the human clinical trials of VRP-based HIV-1 vaccines [146,147].
The use of an optimal delivery carrier for mRNA vaccine entry into the cell is essential for the efficacy of the vaccines. Naked mRNA is prone to degradation and, hence, offers limited efficacy in vivo. However, lipid-based nanoparticles have proven to be successful to deliver mRNA vaccines in humans, as seen in the case of mRNA-1273 and BNT162b as well as remain in clinical trials against various infectious diseases (Table 2). LNPs protect the mRNA from the RNases and help in the effective delivery of mRNA-based vaccines. However, further studies are required to explore the efficacy of other delivery methods, such as CNE, protamine, and VRP in the clinic.

5. Future Perspectives

mRNA vaccine delivery has been effective in preclinical studies and clinical trials; however, there are some challenges that need to be addressed. One such challenge is that during the process of delivery, a large proportion of RNA-loaded carriers becomes trapped in the endosome and gets degraded, thus, decreasing the efficacy [153,154]. Developments that enhance endosomal escape and prevent degradation are highly desirable. Another challenge is targeting the delivery to a specific site in vivo. The current delivery methods induce a plethora of immune cells at the site of injection, which leads to immune stimulation [155,156]. In vivo targeting of B cells, T cells, macrophages and dendritic cells will aid in increasing immunization efficiency [156]. The safety of the delivery vehicles, such as polymers and cationic lipids, remains a concern. These delivery vehicles may induce enhanced membrane fusion, endosomal disruption and cell stress, which can lead to cytotoxicity [157,158]. Therefore, safe delivery materials, such as biodegradable materials or the ones that mask cationic charges, are necessary.
The molecular mechanisms in the delivery process remain to be explored in depth to facilitate the development of effective immunization by mRNA vaccines. A better understanding of the delivery formats, administration routes and carrier materials, as well as pathways responsible for cellular uptake, cytosolic release, endosomal escape, lysosomal degradation and exocytosis is also required.
Given the success of mRNA vaccines in the COVID-19 pandemic, they are a promising alternative to the traditional vaccine platforms. mRNA vaccines are manufactured quickly and designed for emerging infectious diseases [159]. Henceforth, there has been an increase in the intellectual property (IP) landscape for mRNA-based vaccines. A recent report generated a ten-year landscape for mRNA vaccines’ IP. They identified 113 INPADOC patent families and indexed them based on the indication, methods of delivery and pharmacological modifications. It was also observed that patent filing dramatically increased over the past 5 years for cancer and infectious diseases. There were increased patent applications for emerging infectious diseases, such as Ebola virus, Zika virus, MERS-CoV2 and SARS-CoV2. Around 70% of the patents were filed by industry and the remaining were filed by academic institutions or independent investors. There was an increase in patent filing to protect the methods to improve mRNA delivery efficiency, especially for lipid-based nanoparticles, followed by nucleoside modified, sequence or codon-optimized mRNA or poly-A tail modified and self-amplifying mRNA [160].
During the COVID-19 pandemic, LNP-based mRNA vaccines have proven to be a quick and effective vaccination strategy; several other mRNA vaccines against various infectious diseases, such as HIV, Rabies virus, Influenza virus, Zika virus, Ebola virus and cancers, remain in clinical trials. However, there is a need to optimize the safety and increase the efficacy of mRNA vaccines [26]. So far, LNPs have been proven to be an effective delivery method for vaccination against SARS-CoV2 in humans [8]. The improvements in delivery methods and vaccine formulations will make mRNA vaccines an important class of vaccines against emerging infectious diseases and cancers.

Author Contributions

Conceptualization, N., J.W. and A.-M.H.; software, N.; formal analysis, N., J.W. and A.-M.H.; investigation, N., J.W. and A.-M.H.; resources, N., J.W. and A.-M.H.; data curation, N.; writing—original draft preparation, N., J.W. and A.-M.H.; writing—review and editing, N., J.W. and A.-M.H.; visualization, N., J.W. and A.-M.H.; supervision, N., J.W. and A.-M.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The company had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the results.

References

  1. Xu, S.; Yang, K.; Li, R.; Zhang, L. mRNA Vaccine Era-Mechanisms, Drug Platform and Clinical Prospection. Int. J. Mol. Sci. 2020, 21, 6582. [Google Scholar] [CrossRef] [PubMed]
  2. Schlake, T.; Thess, A.; Fotin-Mleczek, M.; Kallen, K.J. Developing mRNA-vaccine technologies. RNA Biol. 2012, 9, 1319–1330. [Google Scholar] [CrossRef] [PubMed]
  3. Kowalski, P.S.; Rudra, A.; Miao, L.; Anderson, D.G. Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol. Ther. 2019, 27, 710–728. [Google Scholar] [CrossRef] [PubMed]
  4. Damase, T.R.; Sukhovershin, R.; Boada, C.; Taraballi, F.; Pettigrew, R.I.; Cooke, J.P. The Limitless Future of RNA Therapeutics. Front. Bioeng. Biotechnol. 2021, 9, 628137. [Google Scholar] [CrossRef]
  5. Watts, J.K.; Corey, D.R. Silencing disease genes in the laboratory and the clinic. J. Pathol. 2012, 226, 365–379. [Google Scholar] [CrossRef] [PubMed]
  6. Hou, X.; Zaks, T.; Langer, R.; Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 2021, 6, 1078–1094. [Google Scholar] [CrossRef]
  7. Wadhwa, A.; Aljabbari, A.; Lokras, A.; Foged, C.; Thakur, A. Opportunities and Challenges in the Delivery of mRNA-based Vaccines. Pharmaceutics 2020, 12, 102. [Google Scholar] [CrossRef] [PubMed]
  8. Wang, Y.; Zhang, Z.; Luo, J.; Han, X.; Wei, Y.; Wei, X. mRNA vaccine: A potential therapeutic strategy. Mol. Cancer 2021, 20, 33. [Google Scholar] [CrossRef]
  9. van den Berg, A.I.S.; Yun, C.-O.; Schiffelers, R.M.; Hennink, W.E. Polymeric delivery systems for nucleic acid therapeutics: Approaching the clinic. J. Control. Release 2021, 331, 121–141. [Google Scholar] [CrossRef]
  10. Reichmuth, A.M.; Oberli, M.A.; Jaklenec, A.; Langer, R.; Blankschtein, D. mRNA vaccine delivery using lipid nanoparticles. Ther. Deliv. 2016, 7, 319–334. [Google Scholar] [CrossRef]
  11. Wei, J.; Hui, A.-M. The Development of mRNA Vaccines for Infectious Diseases: Recent Updates. Infect. Drug. Resist. 2021, 14, 5271–5285. [Google Scholar] [CrossRef]
  12. Aldosari, B.N.; Alfagih, I.M.; Almurshedi, A.S. Lipid Nanoparticles as Delivery Systems for RNA-Based Vaccines. Pharmaceutics 2021, 13, 206. [Google Scholar] [CrossRef] [PubMed]
  13. Miao, L.; Zhang, Y.; Huang, L. mRNA vaccine for cancer immunotherapy. Mol. Cancer 2021, 20, 41. [Google Scholar] [CrossRef] [PubMed]
  14. Fang, E.; Liu, X.; Li, M.; Zhang, Z.; Song, L.; Zhu, B.; Wu, X.; Liu, J.; Zhao, D.; Li, Y. Advances in COVID-19 mRNA vaccine development. Signal Transduct. Target. Ther. 2022, 7, 94. [Google Scholar] [CrossRef]
  15. Dolgin, E. The tangled history of mRNA vaccines. Nature 2021, 597, 318–324. [Google Scholar] [CrossRef]
  16. Park, J.W.; Lagniton, P.N.P.; Liu, Y.; Xu, R.H. mRNA vaccines for COVID-19: What, why and how. Int. J. Biol. Sci. 2021, 17, 1446–1460. [Google Scholar] [CrossRef]
  17. Liu, T.; Liang, Y.; Huang, L. Development and Delivery Systems of mRNA Vaccines. Front. Bioeng. Biotechnol. 2021, 9, 718753. [Google Scholar] [CrossRef]
  18. Sahin, U.; Karikó, K.; Türeci, Ö. mRNA-based therapeutics—Developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef]
  19. Tang, X.; Zhang, S.; Fu, R.; Zhang, L.; Huang, K.; Peng, H.; Dai, L.; Chen, Q. Therapeutic Prospects of mRNA-Based Gene Therapy for Glioblastoma. Front. Oncol. 2019, 9, 1208. [Google Scholar] [CrossRef]
  20. Schlich, M.; Palomba, R.; Costabile, G.; Mizrahy, S.; Pannuzzo, M.; Peer, D.; Decuzzi, P. Cytosolic delivery of nucleic acids: The case of ionizable lipid nanoparticles. Bioeng. Transl. Med. 2021, 6, e10213. [Google Scholar] [CrossRef]
  21. Delehedde, C.; Even, L.; Midoux, P.; Pichon, C.; Perche, F. Intracellular Routing and Recognition of Lipid-Based mRNA Nanoparticles. Pharmaceutics 2021, 13, 945. [Google Scholar] [CrossRef] [PubMed]
  22. Sartorius, R.; Trovato, M.; Manco, R.; D’Apice, L.; De Berardinis, P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines 2021, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  23. El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like receptors activation, signaling, and targeting: An overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef]
  24. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug. Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, C.; Maruggi, G.; Shan, H.; Li, J. Advances in mRNA Vaccines for Infectious Diseases. Front. Immunol. 2019, 10, 594. [Google Scholar] [CrossRef] [PubMed]
  26. Zeng, C.; Zhang, C.; Walker, P.G.; Dong, Y. Formulation and Delivery Technologies for mRNA Vaccines; Springer: Berlin/Heidelberg, Germany, 2020; pp. 1–40. [Google Scholar]
  27. Uddin, M.N.; Allon, A.; Roni, M.A.; Kouzi, S. Overview and Future Potential of Fast Dissolving Buccal Films as Drug Delivery System for Vaccines. J. Pharm. Pharm. Sci. 2019, 22, 388–406. [Google Scholar] [CrossRef]
  28. Pollard, A.J.; Bijker, E.M. A guide to vaccinology: From basic principles to new developments. Nat. Rev. Immunol. 2021, 21, 83–100. [Google Scholar] [CrossRef]
  29. Tiwari, G.; Tiwari, R.; Sriwastawa, B.; Bhati, L.; Pandey, S.; Pandey, P.; Bannerjee, S.K. Drug delivery systems: An updated review. Int. J. Pharm. Investig. 2012, 2, 2–11. [Google Scholar] [CrossRef]
  30. Hervé, C.; Laupèze, B.; Del Giudice, G.; Didierlaurent, A.M.; Tavares Da Silva, F. The how’s and what’s of vaccine reactogenicity. NPJ Vaccines 2019, 4, 39. [Google Scholar] [CrossRef]
  31. Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.-H.; Qoronfleh, M.W. Therapeutic efficacy of nanoparticles and routes of administration. Biomater. Res. 2019, 23, 20. [Google Scholar] [CrossRef]
  32. Jin, J.-F.; Zhu, L.-L.; Chen, M.; Xu, H.-M.; Wang, H.-F.; Feng, X.-Q.; Zhu, X.-P.; Zhou, Q. The optimal choice of medication administration route regarding intravenous, intramuscular, and subcutaneous injection. Patient Prefer. Adherence 2015, 9, 923–942. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, J.; Eygeris, Y.; Gupta, M.; Sahay, G. Self-assembled mRNA vaccines. Adv. Drug. Deliv. Rev. 2021, 170, 83–112. [Google Scholar] [CrossRef]
  34. Homayun, B.; Lin, X.; Choi, H.-J. Challenges and Recent Progress in Oral Drug Delivery Systems for Biopharmaceuticals. Pharmaceutics 2019, 11, 129. [Google Scholar] [CrossRef] [PubMed]
  35. Hettinga, J.; Carlisle, R. Vaccination into the Dermal Compartment: Techniques, Challenges, and Prospects. Vaccines 2020, 8, 534. [Google Scholar] [CrossRef] [PubMed]
  36. Turner, M.R.; Balu-Iyer, S.V. Challenges and Opportunities for the Subcutaneous Delivery of Therapeutic Proteins. J. Pharm. Sci. 2018, 107, 1247–1260. [Google Scholar] [CrossRef] [PubMed]
  37. Usach, I.; Martinez, R.; Festini, T.; Peris, J.-E. Subcutaneous Injection of Drugs: Literature Review of Factors Influencing Pain Sensation at the Injection Site. Adv. Ther. 2019, 36, 2986–2996. [Google Scholar] [CrossRef] [PubMed]
  38. Nicholson, L.B. The immune system. Essays Biochem. 2016, 60, 275–301. [Google Scholar] [CrossRef]
  39. Zuckerman, J.N. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ (Clin. Res. Ed.) 2000, 321, 1237–1238. [Google Scholar] [CrossRef]
  40. Schoenmaker, L.; Witzigmann, D.; Kulkarni, J.A.; Verbeke, R.; Kersten, G.; Jiskoot, W.; Crommelin, D.J.A. mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability. Int. J. Pharm. 2021, 601, 120586. [Google Scholar] [CrossRef]
  41. Turner, J.S.; O’Halloran, J.A.; Kalaidina, E.; Kim, W.; Schmitz, A.J.; Zhou, J.Q.; Lei, T.; Thapa, M.; Chen, R.E.; Case, J.B.; et al. SARS-CoV-2 mRNA vaccines induce persistent human germinal centre responses. Nature 2021, 596, 109–113. [Google Scholar] [CrossRef]
  42. He, Q.; Gao, H.; Tan, D.; Zhang, H.; Wang, J.-Z. mRNA cancer vaccines: Advances, trends and challenges. Acta Pharm. Sin. B 2022, 12, 2969–2989. [Google Scholar] [CrossRef]
  43. Joe, P.T.; Christopoulou, I.; van Hoecke, L.; Schepens, B.; Ysenbaert, T.; Heirman, C.; Thielemans, K.; Saelens, X.; Aerts, J.L. Intranodal administration of mRNA encoding nucleoprotein provides cross-strain immunity against influenza in mice. J. Transl. Med. 2019, 17, 242. [Google Scholar] [CrossRef]
  44. Van Lint, S.; Renmans, D.; Broos, K.; Dewitte, H.; Lentacker, I.; Heirman, C.; Breckpot, K.; Thielemans, K. The ReNAissanCe of mRNA-based cancer therapy. Expert Rev. Vaccines 2015, 14, 235–251. [Google Scholar] [CrossRef]
  45. Sun, X.; Zhou, F.; Bai, X.; Yuan, Q.; Zhang, M.; Ma, L.; Jin, Y. Application of ultrasound-guided intranodal lymphangiography and embolisation in cancer patients with postoperative lymphatic leakage. World J. Surg. Oncol. 2021, 19, 32. [Google Scholar] [CrossRef]
  46. Heida, R.; Hinrichs, W.L.; Frijlink, H.W. Inhaled vaccine delivery in the combat against respiratory viruses: A 2021 overview of recent developments and implications for COVID-19. Expert Rev. Vaccines 2021, 21, 957–974. [Google Scholar] [CrossRef]
  47. Hussain, S.S.; George, S.; Singh, S.; Jayant, R.; Hu, C.A.; Sopori, M.; Chand, H.S. A Small Molecule BH3-mimetic Suppresses Cigarette Smoke-Induced Mucous Expression in Airway Epithelial Cells. Sci. Rep. 2018, 8, 13796. [Google Scholar] [CrossRef]
  48. Chand, H.S.; Vazquez-Guillamet, R.; Royer, C.; Rudolph, K.; Mishra, N.; Singh, S.P.; Hussain, S.S.; Barrett, E.; Callen, S.; Byrareddy, S.N.; et al. Cigarette smoke and HIV synergistically affect lung pathology in cynomolgus macaques. J. Clin. Investig. 2018, 128, 5428–5433. [Google Scholar] [CrossRef]
  49. Silva-Sanchez, A.; Randall, T.D. Role of iBALT in Respiratory Immunity. In Inducible Lymphoid Organs; Kabashima, K., Egawa, G., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 21–43. [Google Scholar]
  50. Diken, M.; Kreiter, S.; Selmi, A.; Britten, C.M.; Huber, C.; Türeci, Ö.; Sahin, U. Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation. Gene Ther. 2011, 18, 702–708. [Google Scholar] [CrossRef]
  51. Selmi, A.; Vascotto, F.; Kautz-Neu, K.; Türeci, Ö.; Sahin, U.; von Stebut, E.; Diken, M.; Kreiter, S. Uptake of synthetic naked RNA by skin-resident dendritic cells via macropinocytosis allows antigen expression and induction of T-cell responses in mice. Cancer Immunol. Immunother. 2016, 65, 1075–1083. [Google Scholar] [CrossRef]
  52. Redka, D.Y.S.; Gütschow, M.; Grinstein, S.; Canton, J. Differential ability of proinflammatory and anti-inflammatory macrophages to perform macropinocytosis. Mol. Biol. Cell 2018, 29, 53–65. [Google Scholar] [CrossRef]
  53. Stewart, M.P.; Langer, R.; Jensen, K.F. Intracellular Delivery by Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem. Rev. 2018, 118, 7409–7531. [Google Scholar] [CrossRef]
  54. Jones, K.L.; Drane, D.; Gowans, E.J. Long-term storage of DNA-free RNA for use in vaccine studies. Biotechniques 2007, 43, 675–681. [Google Scholar] [CrossRef]
  55. Schlee, M.; Hartmann, G. Discriminating self from non-self in nucleic acid sensing. Nat. Rev. Immunol. 2016, 16, 566–580. [Google Scholar] [CrossRef]
  56. Edwards, D.K.; Jasny, E.; Yoon, H.; Horscroft, N.; Schanen, B.; Geter, T.; Fotin-Mleczek, M.; Petsch, B.; Wittman, V. Adjuvant effects of a sequence-engineered mRNA vaccine: Translational profiling demonstrates similar human and murine innate response. J. Transl. Med. 2017, 15, 1. [Google Scholar] [CrossRef]
  57. Anderson, B.R.; Muramatsu, H.; Nallagatla, S.R.; Bevilacqua, P.C.; Sansing, L.H.; Weissman, D.; Karikó, K. Incorporation of pseudouridine into mRNA enhances translation by diminishing PKR activation. Nucleic Acids Res. 2010, 38, 5884–5892. [Google Scholar] [CrossRef]
  58. Singer, D.F.; Linderman, J.J. The relationship between antigen concentration, antigen internalization, and antigenic complexes: Modeling insights into antigen processing and presentation. J. Cell Biol. 1990, 111, 55–68. [Google Scholar] [CrossRef]
  59. Canton, J. Macropinocytosis: New Insights Into Its Underappreciated Role in Innate Immune Cell Surveillance. Front. Immunol. 2018, 9, 2286. [Google Scholar] [CrossRef]
  60. Ying, H.; Zaks, T.Z.; Wang, R.-F.; Irvine, K.R.; Kammula, U.S.; Marincola, F.M.; Leitner, W.W.; Restifo, N.P. Cancer therapy using a self-replicating RNA vaccine. Nat. Med. 1999, 5, 823–827. [Google Scholar] [CrossRef]
  61. Fleeton, M.N.; Chen, M.; Berglund, P.; Rhodes, G.; Parker, S.E.; Murphy, M.; Atkins, G.J.; Liljestrom, P. Self-replicative RNA vaccines elicit protection against influenza A virus, respiratory syncytial virus, and a tickborne encephalitis virus. J. Infect. Dis. 2001, 183, 1395–1398. [Google Scholar] [CrossRef]
  62. Kreiter, S.; Selmi, A.; Diken, M.; Koslowski, M.; Britten, C.M.; Huber, C.; Türeci, Ö.; Sahin, U. Intranodal Vaccination with Naked Antigen-Encoding RNA Elicits Potent Prophylactic and Therapeutic Antitumoral Immunity. Cancer Res. 2010, 70, 9031–9040. [Google Scholar] [CrossRef]
  63. Bialkowski, L.; van Weijnen, A.; Van der Jeught, K.; Renmans, D.; Daszkiewicz, L.; Heirman, C.; Stangé, G.; Breckpot, K.; Aerts, J.L.; Thielemans, K. Intralymphatic mRNA vaccine induces CD8 T-cell responses that inhibit the growth of mucosally located tumours. Sci. Rep. 2016, 6, 22509. [Google Scholar] [CrossRef] [PubMed]
  64. Tiwari, P.M.; Vanover, D.; Lindsay, K.E.; Bawage, S.S.; Kirschman, J.L.; Bhosle, S.; Lifland, A.W.; Zurla, C.; Santangelo, P.J. Engineered mRNA-expressed antibodies prevent respiratory syncytial virus infection. Nat. Commun. 2018, 9, 3999. [Google Scholar] [CrossRef] [PubMed]
  65. Lorenzi, J.C.C.; Trombone, A.P.F.; Rocha, C.D.; Almeida, L.P.; Lousada, R.L.; Malardo, T.; Fontoura, I.C.; Rossetti, R.A.M.; Gembre, A.F.; Silva, A.M.; et al. Intranasal vaccination with messenger RNA as a new approach in gene therapy: Use against tuberculosis. BMC Biotechnol. 2010, 10, 77. [Google Scholar] [CrossRef]
  66. Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.-P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef]
  67. Leal, L.; Guardo, A.C.; Morón-López, S.; Salgado, M.; Mothe, B.; Heirman, C.; Pannus, P.; Vanham, G.; van den Ham, H.J.; Gruters, R.; et al. Phase I clinical trial of an intranodally administered mRNA-based therapeutic vaccine against HIV-1 infection. AIDS (Lond. Engl.) 2018, 32, 2533–2545. [Google Scholar] [CrossRef] [PubMed]
  68. Gaudino, S.J.; Kumar, P. Cross-Talk Between Antigen Presenting Cells and T Cells Impacts Intestinal Homeostasis, Bacterial Infections, and Tumorigenesis. Front. Immunol. 2019, 10, 360. [Google Scholar] [CrossRef]
  69. Eiz-Vesper, B.; Schmetzer, H.M. Antigen-Presenting Cells: Potential of Proven und New Players in Immune Therapies. Transfus. Med. Hemother. 2020, 47, 429–431. [Google Scholar] [CrossRef]
  70. Mbongue, J.; Nicholas, D.; Firek, A.; Langridge, W. The role of dendritic cells in tissue-specific autoimmunity. J. Immunol. Res. 2014, 2014, 857143. [Google Scholar] [CrossRef]
  71. Garg, A.D.; Coulie, P.G.; Van den Eynde, B.J.; Agostinis, P. Integrating Next-Generation Dendritic Cell Vaccines into the Current Cancer Immunotherapy Landscape. Trends Immunol. 2017, 38, 577–593. [Google Scholar] [CrossRef]
  72. Sherba, J.J.; Hogquist, S.; Lin, H.; Shan, J.W.; Shreiber, D.I.; Zahn, J.D. The effects of electroporation buffer composition on cell viability and electro-transfection efficiency. Sci. Rep. 2020, 10, 3053. [Google Scholar] [CrossRef]
  73. Lee, D.; Chan, S.S.Y.; Aksic, N.; Bajalovic, N.; Loke, D.K. Ultralong-Time Recovery and Low-Voltage Electroporation for Biological Cell Monitoring Enabled by a Microsized Multipulse Framework. ACS Omega 2021, 6, 35325–35333. [Google Scholar] [CrossRef] [PubMed]
  74. Wilgenhof, S.; Van Nuffel, A.M.T.; Benteyn, D.; Corthals, J.; Aerts, C.; Heirman, C.; Van Riet, I.; Bonehill, A.; Thielemans, K.; Neyns, B. A phase IB study on intravenous synthetic mRNA electroporated dendritic cell immunotherapy in pretreated advanced melanoma patients. Ann. Oncol. 2013, 24, 2686–2693. [Google Scholar] [CrossRef] [PubMed]
  75. Mitchell, D.A.; Batich, K.A.; Gunn, M.D.; Huang, M.-N.; Sanchez-Perez, L.; Nair, S.K.; Congdon, K.L.; Reap, E.A.; Archer, G.E.; Desjardins, A.; et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature 2015, 519, 366–369. [Google Scholar] [CrossRef] [PubMed]
  76. Batich, K.A.; Reap, E.A.; Archer, G.E.; Sanchez-Perez, L.; Nair, S.K.; Schmittling, R.J.; Norberg, P.; Xie, W.; Herndon, J.E., II; Healy, P.; et al. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clin. Cancer Res. 2017, 23, 1898–1909. [Google Scholar] [CrossRef]
  77. Reche, P.; Flower, D.R.; Fridkis-Hareli, M.; Hoshino, Y. Peptide-Based Immunotherapeutics and Vaccines 2017. J. Immunol. Res. 2018, 2018, 4568239. [Google Scholar] [CrossRef]
  78. Li, W.; Joshi, M.D.; Singhania, S.; Ramsey, K.H.; Murthy, A.K. Peptide Vaccine: Progress and Challenges. Vaccines 2014, 2, 515–536. [Google Scholar] [CrossRef]
  79. Grau, M.; Walker, P.R.; Derouazi, M. Mechanistic insights into the efficacy of cell penetrating peptide-based cancer vaccines. Cell. Mol. Life Sci. 2018, 75, 2887–2896. [Google Scholar] [CrossRef]
  80. Qiu, Y.; Man, R.C.H.; Liao, Q.; Kung, K.L.K.; Chow, M.Y.T.; Lam, J.K.W. Effective mRNA pulmonary delivery by dry powder formulation of PEGylated synthetic KL4 peptide. J. Control. Release 2019, 314, 102–115. [Google Scholar] [CrossRef]
  81. Udhayakumar, V.K.; De Beuckelaer, A.; McCaffrey, J.; McCrudden, C.M.; Kirschman, J.L.; Vanover, D.; Van Hoecke, L.; Roose, K.; Deswarte, K.; De Geest, B.G.; et al. Arginine-Rich Peptide-Based mRNA Nanocomplexes Efficiently Instigate Cytotoxic T Cell Immunity Dependent on the Amphipathic Organization of the Peptide. Adv. Healthc. Mater. 2017, 6, 1601412. [Google Scholar] [CrossRef]
  82. Alberer, M.; Gnad-Vogt, U.; Hong, H.S.; Mehr, K.T.; Backert, L.; Finak, G.; Gottardo, R.; Bica, M.A.; Garofano, A.; Koch, S.D.; et al. Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: An open-label, non-randomised, prospective, first-in-human phase 1 clinical trial. Lancet 2017, 390, 1511–1520. [Google Scholar] [CrossRef]
  83. Stitz, L.; Vogel, A.; Schnee, M.; Voss, D.; Rauch, S.; Mutzke, T.; Ketterer, T.; Kramps, T.; Petsch, B. A thermostable messenger RNA based vaccine against rabies. PLoS Negl. Trop. Dis. 2017, 11, e0006108. [Google Scholar] [CrossRef] [PubMed]
  84. Scheel, B.; Teufel, R.; Probst, J.; Carralot, J.-P.; Geginat, J.; Radsak, M.; Jarrossay, D.; Wagner, H.; Jung, G.; Rammensee, H.-G.; et al. Toll-like receptor-dependent activation of several human blood cell types by protamine-condensed mRNA. Eur. J. Immunol. 2005, 35, 1557–1566. [Google Scholar] [CrossRef]
  85. Fotin-Mleczek, M.; Duchardt, K.M.; Lorenz, C.; Pfeiffer, R.; Ojkic-Zrna, S.; Probst, J.; Kallen, K.J. Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and provide antitumor activity. J. Immunother. 2011, 34, 1–15. [Google Scholar] [CrossRef] [PubMed]
  86. Scheel, B.; Aulwurm, S.; Probst, J.; Stitz, L.; Hoerr, I.; Rammensee, H.-G.; Weller, M.; Pascolo, S. Therapeutic anti-tumor immunity triggered by injections of immunostimulating single-stranded RNA. Eur. J. Immunol. 2006, 36, 2807–2816. [Google Scholar] [CrossRef]
  87. Scheel, B.; Braedel, S.; Probst, J.; Carralot, J.-P.; Wagner, H.; Schild, H.; Jung, G.; Rammensee, H.-G.; Pascolo, S. Immunostimulating capacities of stabilized RNA molecules. Eur. J. Immunol. 2004, 34, 537–547. [Google Scholar] [CrossRef]
  88. Dong, Y.; Dorkin, J.R.; Wang, W.; Chang, P.H.; Webber, M.J.; Tang, B.C.; Yang, J.; Abutbul-Ionita, I.; Danino, D.; DeRosa, F.; et al. Poly(glycoamidoamine) Brushes Formulated Nanomaterials for Systemic siRNA and mRNA Delivery in Vivo. Nano Lett. 2016, 16, 842–848. [Google Scholar] [CrossRef] [PubMed]
  89. Patel, A.K.; Kaczmarek, J.C.; Bose, S.; Kauffman, K.J.; Mir, F.; Heartlein, M.W.; DeRosa, F.; Langer, R.; Anderson, D.G. Inhaled Nanoformulated mRNA Polyplexes for Protein Production in Lung Epithelium. Adv. Mater. 2019, 31, 1805116. [Google Scholar] [CrossRef] [PubMed]
  90. McCullough, K.C.; Bassi, I.; Milona, P.; Suter, R.; Thomann-Harwood, L.; Englezou, P.; Demoulins, T.; Ruggli, N. Self-replicating Replicon-RNA Delivery to Dendritic Cells by Chitosan-nanoparticles for Translation In Vitro and In Vivo. Mol. Nucleic Acids 2014, 3, e173. [Google Scholar] [CrossRef]
  91. Chahal, J.S.; Khan, O.F.; Cooper, C.L.; McPartlan, J.S.; Tsosie, J.K.; Tilley, L.D.; Sidik, S.M.; Lourido, S.; Langer, R.; Bavari, S.; et al. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single dose. Proc. Natl. Acad. Sci. USA 2016, 113, E4133–E4142. [Google Scholar] [CrossRef]
  92. Li, W.; Ma, L.; Guo, L.-P.; Wang, X.-L.; Zhang, J.-W.; Bu, Z.-G.; Hua, R.-H. West Nile virus infectious replicon particles generated using a packaging-restricted cell line is a safe reporter system. Sci. Rep. 2017, 7, 3286. [Google Scholar] [CrossRef]
  93. Moyo, N.; Vogel, A.B.; Buus, S.; Erbar, S.; Wee, E.G.; Sahin, U.; Hanke, T. Efficient Induction of T Cells against Conserved HIV-1 Regions by Mosaic Vaccines Delivered as Self-Amplifying mRNA. Mol. Ther. Methods Clin. Dev. 2019, 12, 32–46. [Google Scholar] [CrossRef] [PubMed]
  94. Chowdhury, S.; Toth, I.; Stephenson, R.J. Dendrimers in vaccine delivery: Recent progress and advances. Biomaterials 2022, 280, 121303. [Google Scholar] [CrossRef] [PubMed]
  95. Ishaqat, A.; Herrmann, A. Polymers Strive for Accuracy: From Sequence-Defined Polymers to mRNA Vaccines against COVID-19 and Polymers in Nucleic Acid Therapeutics. J. Am. Chem. Soc. 2021, 143, 20529–20545. [Google Scholar] [CrossRef]
  96. Corthésy, B.; Bioley, G. Lipid-Based Particles: Versatile Delivery Systems for Mucosal Vaccination against Infection. Front. Immunol. 2018, 9, 431. [Google Scholar] [CrossRef] [PubMed]
  97. Ganesan, P.; Narayanasamy, D. Lipid nanoparticles: Different preparation techniques, characterization, hurdles, and strategies for the production of solid lipid nanoparticles and nanostructured lipid carriers for oral drug delivery. Sustain. Chem. Pharm. 2017, 6, 37–56. [Google Scholar] [CrossRef]
  98. Maugeri, M.; Nawaz, M.; Papadimitriou, A.; Angerfors, A.; Camponeschi, A.; Na, M.; Hölttä, M.; Skantze, P.; Johansson, S.; Sundqvist, M.; et al. Linkage between endosomal escape of LNP-mRNA and loading into EVs for transport to other cells. Nat. Commun. 2019, 10, 4333. [Google Scholar] [CrossRef]
  99. Zhang, J.; Shrivastava, S.; Cleveland, R.O.; Rabbitts, T.H. Lipid-mRNA Nanoparticle Designed to Enhance Intracellular Delivery Mediated by Shock Waves. ACS Appl. Mater. Interfaces 2019, 11, 10481–10491. [Google Scholar] [CrossRef]
  100. Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Dewitte, H. Three decades of messenger RNA vaccine development. Nano Today 2019, 28, 100766. [Google Scholar] [CrossRef]
  101. Semple, S.C.; Akinc, A.; Chen, J.; Sandhu, A.P.; Mui, B.L.; Cho, C.K.; Sah, D.W.Y.; Stebbing, D.; Crosley, E.J.; Yaworski, E.; et al. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 2010, 28, 172–176. [Google Scholar] [CrossRef]
  102. Billingsley, M.M.; Singh, N.; Ravikumar, P.; Zhang, R.; June, C.H.; Mitchell, M.J. Ionizable Lipid Nanoparticle-Mediated mRNA Delivery for Human CAR T Cell Engineering. Nano Lett. 2020, 20, 1578–1589. [Google Scholar] [CrossRef]
  103. Cullis, P.R.; Hope, M.J. Lipid Nanoparticle Systems for Enabling Gene Therapies. Mol. Ther. 2017, 25, 1467–1475. [Google Scholar] [CrossRef] [PubMed]
  104. Yingchoncharoen, P.; Kalinowski, D.S.; Richardson, D.R. Lipid-Based Drug Delivery Systems in Cancer Therapy: What Is Available and What Is Yet to Come. Pharm. Rev. 2016, 68, 701–787. [Google Scholar] [CrossRef] [PubMed]
  105. Ambegia, E.; Ansell, S.; Cullis, P.; Heyes, J.; Palmer, L.; MacLachlan, I. Stabilized plasmid–lipid particles containing PEG-diacylglycerols exhibit extended circulation lifetimes and tumor selective gene expression. Biochim. Biophys. Acta Biomembr. 2005, 1669, 155–163. [Google Scholar] [CrossRef] [PubMed]
  106. Heyes, J.; Palmer, L.; Bremner, K.; MacLachlan, I. Cationic lipid saturation influences intracellular delivery of encapsulated nucleic acids. J. Control. Release 2005, 107, 276–287. [Google Scholar] [CrossRef]
  107. Cheng, X.; Lee, R.J. The role of helper lipids in lipid nanoparticles (LNPs) designed for oligonucleotide delivery. Adv. Drug Deliv. Rev. 2016, 99, 129–137. [Google Scholar] [CrossRef]
  108. Kuehn, B.M. Rare PEG Allergy Triggered Postvaccination Anaphylaxis. JAMA 2021, 325, 1931. [Google Scholar] [CrossRef]
  109. Miao, L.; Li, L.; Huang, Y.; Delcassian, D.; Chahal, J.; Han, J.; Shi, Y.; Sadtler, K.; Gao, W.; Lin, J.; et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat. Biotechnol. 2019, 37, 1174–1185. [Google Scholar] [CrossRef]
  110. Pardi, N.; Tuyishime, S.; Muramatsu, H.; Kariko, K.; Mui, B.L.; Tam, Y.K.; Madden, T.D.; Hope, M.J.; Weissman, D. Expression kinetics of nucleoside-modified mRNA delivered in lipid nanoparticles to mice by various routes. J. Control. Release 2015, 217, 345–351. [Google Scholar] [CrossRef]
  111. Chaudhary, N.; Weissman, D.; Whitehead, K.A. mRNA vaccines for infectious diseases: Principles, delivery and clinical translation. Nat. Rev. Drug Discov. 2021, 20, 817–838. [Google Scholar] [CrossRef]
  112. Ganta, S.; Talekar, M.; Singh, A.; Coleman, T.P.; Amiji, M.M. Nanoemulsions in translational research-opportunities and challenges in targeted cancer therapy. AAPS PharmSciTech 2014, 15, 694–708. [Google Scholar] [CrossRef]
  113. Vesikari, T.; Forstén, A.; Herbinger, K.-H.; Cioppa, G.D.; Beygo, J.; Borkowski, A.; Groth, N.; Bennati, M.; von Sonnenburg, F. Safety and immunogenicity of an MF59®-adjuvanted A/H5N1 pre-pandemic influenza vaccine in adults and the elderly. Vaccine 2012, 30, 1388–1396. [Google Scholar] [CrossRef] [PubMed]
  114. Calabro, S.; Tritto, E.; Pezzotti, A.; Taccone, M.; Muzzi, A.; Bertholet, S.; De Gregorio, E.; O’Hagan, D.T.; Baudner, B.; Seubert, A. The adjuvant effect of MF59 is due to the oil-in-water emulsion formulation, none of the individual components induce a comparable adjuvant effect. Vaccine 2013, 31, 3363–3369. [Google Scholar] [CrossRef] [PubMed]
  115. O’Hagan, D.T.; Ott, G.S.; De Gregorio, E.; Seubert, A. The mechanism of action of MF59—An innately attractive adjuvant formulation. Vaccine 2012, 30, 4341–4348. [Google Scholar] [CrossRef] [PubMed]
  116. Seubert, A.; Calabro, S.; Santini, L.; Galli, B.; Genovese, A.; Valentini, S.; Aprea, S.; Colaprico, A.; D’Oro, U.; Giuliani, M.M.; et al. Adjuvanticity of the oil-in-water emulsion MF59 is independent of Nlrp3 inflammasome but requires the adaptor protein MyD88. Proc. Natl. Acad. Sci. USA 2011, 108, 11169–11174. [Google Scholar] [CrossRef]
  117. Brito, L.A.; Chan, M.; Shaw, C.A.; Hekele, A.; Carsillo, T.; Schaefer, M.; Archer, J.; Seubert, A.; Otten, G.R.; Beard, C.W.; et al. A Cationic Nanoemulsion for the Delivery of Next-generation RNA Vaccines. Mol. Ther. 2014, 22, 2118–2129. [Google Scholar] [CrossRef]
  118. Brazzoli, M.; Magini, D.; Bonci, A.; Buccato, S.; Giovani, C.; Kratzer, R.; Zurli, V.; Mangiavacchi, S.; Casini, D.; Brito, L.M.; et al. Induction of Broad-Based Immunity and Protective Efficacy by Self-amplifying mRNA Vaccines Encoding Influenza Virus Hemagglutinin. J. Virol. 2016, 90, 332–344. [Google Scholar] [CrossRef]
  119. Maruggi, G.; Chiarot, E.; Giovani, C.; Buccato, S.; Bonacci, S.; Frigimelica, E.; Margarit, I.; Geall, A.; Bensi, G.; Maione, D. Immunogenicity and protective efficacy induced by self-amplifying mRNA vaccines encoding bacterial antigens. Vaccine 2017, 35, 361–368. [Google Scholar] [CrossRef]
  120. Samsa, M.M.; Dupuy, L.C.; Beard, C.W.; Six, C.M.; Schmaljohn, C.S.; Mason, P.W.; Geall, A.J.; Ulmer, J.B.; Yu, D. Self-Amplifying RNA Vaccines for Venezuelan Equine Encephalitis Virus Induce Robust Protective Immunogenicity in Mice. Mol. Ther. 2019, 27, 850–865. [Google Scholar] [CrossRef]
  121. Chahal, J.S.; Fang, T.; Woodham, A.W.; Khan, O.F.; Ling, J.; Anderson, D.G.; Ploegh, H.L. An RNA nanoparticle vaccine against Zika virus elicits antibody and CD8+ T cell responses in a mouse model. Sci. Rep. 2017, 7, 252. [Google Scholar] [CrossRef]
  122. Bahl, K.; Senn, J.J.; Yuzhakov, O.; Bulychev, A.; Brito, L.A.; Hassett, K.J.; Laska, M.E.; Smith, M.; Almarsson, O.; Thompson, J.; et al. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol. Ther. 2017, 25, 1316–1327. [Google Scholar] [CrossRef]
  123. Kübler, H.; Scheel, B.; Gnad-Vogt, U.; Miller, K.; Schultze-Seemann, W.; Vom Dorp, F.; Parmiani, G.; Hampel, C.; Wedel, S.; Trojan, L.; et al. Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: A first-in-man phase I/IIa study. J. Immunother. Cancer 2015, 3, 26. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, Y.; Deng, L.; Kang, S.-M.; Wang, B.-Z. Universal influenza vaccines: From viruses to nanoparticles. Expert. Rev. Vaccines 2018, 17, 967–976. [Google Scholar] [CrossRef] [PubMed]
  125. Meyer, M.; Huang, E.; Yuzhakov, O.; Ramanathan, P.; Ciaramella, G.; Bukreyev, A. Modified mRNA-Based Vaccines Elicit Robust Immune Responses and Protect Guinea Pigs From Ebola Virus Disease. J. Infect. Dis. 2018, 217, 451–455. [Google Scholar] [CrossRef] [PubMed]
  126. Melo, M.; Porter, E.; Zhang, Y.; Silva, M.; Li, N.; Dobosh, B.; Liguori, A.; Skog, P.; Landais, E.; Menis, S.; et al. Immunogenicity of RNA Replicons Encoding HIV Env Immunogens Designed for Self-Assembly into Nanoparticles. Mol. Ther. J. Am. Soc. Gene Ther. 2019, 27, 2080–2090. [Google Scholar] [CrossRef]
  127. Pardi, N.; Secreto, A.J.; Shan, X.; Debonera, F.; Glover, J.; Yi, Y.; Muramatsu, H.; Ni, H.; Mui, B.L.; Tam, Y.K.; et al. Administration of nucleoside-modified mRNA encoding broadly neutralizing antibody protects humanized mice from HIV-1 challenge. Nat. Commun. 2017, 8, 14630. [Google Scholar] [CrossRef]
  128. Anderson, E.J.; Rouphael, N.G.; Widge, A.T.; Jackson, L.A.; Roberts, P.C.; Makhene, M.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; Pruijssers, A.J.; et al. Safety and Immunogenicity of SARS-CoV-2 mRNA-1273 Vaccine in Older Adults. N. Engl. J. Med. 2020, 383, 2427–2438. [Google Scholar] [CrossRef]
  129. Lamb, Y.N. BNT162b2 mRNA COVID-19 Vaccine: First Approval. Drugs 2021, 81, 495–501. [Google Scholar] [CrossRef]
  130. Zhao, M.; Li, M.; Zhang, Z.; Gong, T.; Sun, X. Induction of HIV-1 gag specific immune responses by cationic micelles mediated delivery of gag mRNA. Drug. Deliv. 2016, 23, 2596–2607. [Google Scholar] [CrossRef]
  131. White, L.J.; Sariol, C.A.; Mattocks, M.D.; Wahala, M.P.B.W.; Yingsiwaphat, V.; Collier, M.L.; Whitley, J.; Mikkelsen, R.; Rodriguez, I.V.; Martinez, M.I.; et al. An Alphavirus Vector-Based Tetravalent Dengue Vaccine Induces a Rapid and Protective Immune Response in Macaques That Differs Qualitatively from Immunity Induced by Live Virus Infection. J. Virol. 2013, 87, 3409–3424. [Google Scholar] [CrossRef]
  132. Vander Veen, R.L.; Loynachan, A.T.; Mogler, M.A.; Russell, B.J.; Harris, D.L.; Kamrud, K.I. Safety, immunogenicity, and efficacy of an alphavirus replicon-based swine influenza virus hemagglutinin vaccine. Vaccine 2012, 30, 1944–1950. [Google Scholar] [CrossRef]
  133. Brand, D.; Lemiale, F.; Turbica, I.; Buzelay, L.; Brunet, S.; Barin, F. Comparative analysis of humoral immune responses to HIV type 1 envelope glycoproteins in mice immunized with a DNA vaccine, recombinant Semliki Forest virus RNA, or recombinant Semliki Forest virus particles. AIDS Res. Hum. Retrovir. 1998, 14, 1369–1377. [Google Scholar] [CrossRef] [PubMed]
  134. Pyankov, O.V.; Bodnev, S.A.; Pyankova, O.G.; Solodkyi, V.V.; Pyankov, S.A.; Setoh, Y.X.; Volchkova, V.A.; Suhrbier, A.; Volchkov, V.V.; Agafonov, A.A.; et al. A Kunjin Replicon Virus-like Particle Vaccine Provides Protection Against Ebola Virus Infection in Nonhuman Primates. J. Infect. Dis. 2015, 212 (Suppl. 2), S368–S371. [Google Scholar] [CrossRef] [PubMed]
  135. Sheahan, T.; Whitmore, A.; Long, K.; Ferris, M.; Rockx, B.; Funkhouser, W.; Donaldson, E.; Gralinski, L.; Collier, M.; Heise, M.; et al. Successful vaccination strategies that protect aged mice from lethal challenge from influenza virus and heterologous severe acute respiratory syndrome coronavirus. J. Virol. 2011, 85, 217–230. [Google Scholar] [CrossRef] [PubMed]
  136. Malczyk, A.H.; Kupke, A.; Prüfer, S.; Scheuplein, V.A.; Hutzler, S.; Kreuz, D.; Beissert, T.; Bauer, S.; Hubich-Rau, S.; Tondera, C.; et al. A Highly Immunogenic and Protective Middle East Respiratory Syndrome Coronavirus Vaccine Based on a Recombinant Measles Virus Vaccine Platform. J. Virol. 2015, 89, 11654–11667. [Google Scholar] [CrossRef] [PubMed]
  137. Bates, J.T.; Pickens, J.A.; Schuster, J.E.; Johnson, M.; Tollefson, S.J.; Williams, J.V.; Davis, N.L.; Johnston, R.E.; Schultz-Darken, N.; Slaughter, J.C.; et al. Immunogenicity and efficacy of alphavirus-derived replicon vaccines for respiratory syncytial virus and human metapneumovirus in nonhuman primates. Vaccine 2016, 34, 950–956. [Google Scholar] [CrossRef]
  138. Bernstein, D.I.; Reap, E.A.; Katen, K.; Watson, A.; Smith, K.; Norberg, P.; Olmsted, R.A.; Hoeper, A.; Morris, J.; Negri, S.; et al. Randomized, double-blind, Phase 1 trial of an alphavirus replicon vaccine for cytomegalovirus in CMV seronegative adult volunteers. Vaccine 2009, 28, 484–493. [Google Scholar] [CrossRef]
  139. Lundstrom, K. Replicon RNA Viral Vectors as Vaccines. Vaccines 2016, 4, 39. [Google Scholar] [CrossRef]
  140. Harvey, T.J.; Liu, W.J.; Wang, X.J.; Linedale, R.; Jacobs, M.; Davidson, A.; Le, T.T.T.; Anraku, I.; Suhrbier, A.; Shi, P.-Y.; et al. Tetracycline-Inducible Packaging Cell Line for Production of Flavivirus Replicon Particles. J. Virol. 2004, 78, 531–538. [Google Scholar] [CrossRef]
  141. Usme-Ciro, J.A.; Campillo-Pedroza, N.; Almazán, F.; Gallego-Gomez, J.C. Cytoplasmic RNA viruses as potential vehicles for the delivery of therapeutic small RNAs. Virol. J. 2013, 10, 185. [Google Scholar] [CrossRef]
  142. Vázquez-Calvo, Á.; Saiz, J.-C.; McCullough, K.C.; Sobrino, F.; Martín-Acebes, M.A. Acid-dependent viral entry. Virus Res. 2012, 167, 125–137. [Google Scholar] [CrossRef]
  143. Hoang-Le, D.; Smeenk, L.; Anraku, I.; Pijlman, G.P.; Wang, X.J.; de Vrij, J.; Liu, W.J.; Le, T.T.; Schroder, W.A.; Khromykh, A.A.; et al. A Kunjin replicon vector encoding granulocyte macrophage colony-stimulating factor for intra-tumoral gene therapy. Gene Ther. 2009, 16, 190–199. [Google Scholar] [CrossRef] [PubMed]
  144. Morrison, J.; Plotkin, S. Chapter 19—Viral Vaccines: Fighting Viruses with Vaccines. In Viral Pathogenesis, 3rd ed.; Katze, M.G., Korth, M.J., Law, G.L., Nathanson, N., Eds.; Academic Press: Boston, MA, USA, 2016; pp. 253–269. [Google Scholar] [CrossRef]
  145. Rauch, S.; Jasny, E.; Schmidt, K.E.; Petsch, B. New Vaccine Technologies to Combat Outbreak Situations. Front. Immunol. 2018, 9, 1963. [Google Scholar] [CrossRef] [PubMed]
  146. Wecker, M.; Gilbert, P.; Russell, N.; Hural, J.; Allen, M.; Pensiero, M.; Chulay, J.; Chiu, Y.-L.; Karim, S.S.A.; Burke, D.S. Phase I Safety and Immunogenicity Evaluations of an Alphavirus Replicon HIV-1 Subtype C gag Vaccine in Healthy HIV-1-Uninfected Adults. Clin. Vaccine Immunol. 2012, 19, 1651–1660. [Google Scholar] [CrossRef] [PubMed]
  147. Fuchs, J.D.; Frank, I.; Elizaga, M.L.; Allen, M.; Frahm, N.; Kochar, N.; Li, S.; Edupuganti, S.; Kalams, S.A.; Tomaras, G.D.; et al. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Vesicular Stomatitis Virus Human Immunodeficiency Virus-1 gag Vaccine (HVTN 090). In Open Forum Infectious Diseases; Oxford University Press: Oxford, UK, 2015; Volume 2, p. ofv082. [Google Scholar] [CrossRef]
  148. Mahase, E. COVID-19: Moderna applies for US and EU approval as vaccine trial reports 94.1% efficacy. BMJ 2020, 371, m4709. [Google Scholar] [CrossRef]
  149. Gebre, M.S.; Rauch, S.; Roth, N.; Yu, J.; Chandrashekar, A.; Mercado, N.B.; He, X.; Liu, J.; McMahan, K.; Martinot, A.; et al. Optimization of non-coding regions for a non-modified mRNA COVID-19 vaccine. Nature 2021, 601, 410–414. [Google Scholar] [CrossRef]
  150. Armbruster, N.; Jasny, E.; Petsch, B. Advances in RNA Vaccines for Preventive Indications: A Case Study of A Vaccine Against Rabies. Vaccines 2019, 7, 132. [Google Scholar] [CrossRef]
  151. Aldrich, C.; Leroux-Roels, I.; Huang, K.B.; Bica, M.A.; Loeliger, E.; Schoenborn-Kellenberger, O.; Walz, L.; Leroux-Roels, G.; von Sonnenburg, F.; Oostvogels, L. 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, 39, 1310–1318. [Google Scholar] [CrossRef]
  152. Feldman, R.A.; Fuhr, R.; Smolenov, I.; Mick Ribeiro, A.; Panther, L.; Watson, M.; Senn, J.J.; Smith, M.; Almarsson, Ӧ.; Pujar, H.S.; et al. mRNA vaccines against H10N8 and H7N9 influenza viruses of pandemic potential are immunogenic and well tolerated in healthy adults in phase 1 randomized clinical trials. Vaccine 2019, 37, 3326–3334. [Google Scholar] [CrossRef]
  153. Sahay, G.; Querbes, W.; Alabi, C.; Eltoukhy, A.; Sarkar, S.; Zurenko, C.; Karagiannis, E.; Love, K.; Chen, D.; Zoncu, R.; et al. Efficiency of siRNA delivery by lipid nanoparticles is limited by endocytic recycling. Nat. Biotechnol. 2013, 31, 653–658. [Google Scholar] [CrossRef]
  154. Sayers, E.J.; Peel, S.E.; Schantz, A.; England, R.M.; Beano, M.; Bates, S.M.; Desai, A.S.; Puri, S.; Ashford, M.B.; Jones, A.T. Endocytic Profiling of Cancer Cell Models Reveals Critical Factors Influencing LNP-Mediated mRNA Delivery and Protein Expression. Mol. Ther. 2019, 27, 1950–1962. [Google Scholar] [CrossRef]
  155. Veiga, N.; Goldsmith, M.; Granot, Y.; Rosenblum, D.; Dammes, N.; Kedmi, R.; Ramishetti, S.; Peer, D. Cell specific delivery of modified mRNA expressing therapeutic proteins to leukocytes. Nat. Commun. 2018, 9, 4493. [Google Scholar] [CrossRef] [PubMed]
  156. Fenton, O.S.; Kauffman, K.J.; Kaczmarek, J.C.; McClellan, R.L.; Jhunjhunwala, S.; Tibbitt, M.W.; Zeng, M.D.; Appel, E.A.; Dorkin, J.R.; Mir, F.F.; et al. Synthesis and Biological Evaluation of Ionizable Lipid Materials for the In Vivo Delivery of Messenger RNA to B Lymphocytes. Adv. Mater. 2017, 29, 1606944. [Google Scholar] [CrossRef] [PubMed]
  157. Lv, H.; Zhang, S.; Wang, B.; Cui, S.; Yan, J. Toxicity of cationic lipids and cationic polymers in gene delivery. J. Control. Release 2006, 114, 100–109. [Google Scholar] [CrossRef] [PubMed]
  158. Xue, H.Y.; Liu, S.; Wong, H.L. Nanotoxicity: A key obstacle to clinical translation of siRNA-based nanomedicine. Nanomedicine 2014, 9, 295–312. [Google Scholar] [CrossRef] [PubMed]
  159. Szabó, G.T.; Mahiny, A.J.; Vlatkovic, I. COVID-19 mRNA vaccines: Platforms and current developments. Mol. Ther. 2022, 30, 1850–1868. [Google Scholar] [CrossRef] [PubMed]
  160. Lowery, C.M.D. mRNA vaccines: Intellectual property landscape. Nat. Rev. Drug Discov. 2020, 19, 578–579. [Google Scholar]
Figure 1. The routes of delivery for mRNA vaccines.
Figure 1. The routes of delivery for mRNA vaccines.
Life 12 01254 g001
Figure 2. (A) Delivery carriers for mRNA vaccines and (B) the mechanism of mRNA delivery into the cell.
Figure 2. (A) Delivery carriers for mRNA vaccines and (B) the mechanism of mRNA delivery into the cell.
Life 12 01254 g002
Table 2. mRNA vaccines against infectious diseases in clinic.
Table 2. mRNA vaccines against infectious diseases in clinic.
DiseaseVaccine NameCompanyRoute of AdministrationPhasePlatformReference
mRNA-1273ModernaIMApprovedLNP[148]
BNT162bPfizer/BioNTech/Fosun PharmaIMApprovedLNP[129]
CVnCoVCureVacIMIIb/IIILNPNCT04652102 [149]
SARS-CoV2LUNAR-COV19Arcturus TxIMIILNPNCT04668339
LNP-nCoVsaRNAImperial College London/VacEquity Global HealthIMILNPNCT04934111
ARCoVAcademy of Military Science/Walvax Biotech/SuzhouIMIIILNPNCT04847102
ChulaCoV19Chulalongkorn UniversityIMIILNPNCT04566276
RabiesCV7201CureVacIMILNP[150]
CV7202CureVacIMILNP[151]
InfluenzamRNA-H10N8, mRNA-H7N9ModernaIMILNP[152]
Respiratory syncytial virus (RSV)mRNA-1345ModernaIMILNPNCT04528719
Human metapneumovirus (HMPV) and parainfluenza virus type 3 (PIV3)mRNA-1653ModernaIMIbLNPNCT04144348 NCT03392389
Human Cytomegalovirus (HCMV)mRNA-1647ModernaIMIIILNPNCT05085366
Zika virusmRNA-1893ModernaIMIILNPNCT04917861
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nitika; Wei, J.; Hui, A.-M. The Delivery of mRNA Vaccines for Therapeutics. Life 2022, 12, 1254. https://doi.org/10.3390/life12081254

AMA Style

Nitika, Wei J, Hui A-M. The Delivery of mRNA Vaccines for Therapeutics. Life. 2022; 12(8):1254. https://doi.org/10.3390/life12081254

Chicago/Turabian Style

Nitika, Jiao Wei, and Ai-Min Hui. 2022. "The Delivery of mRNA Vaccines for Therapeutics" Life 12, no. 8: 1254. https://doi.org/10.3390/life12081254

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop