RNA vaccine

Last updated

mRNA in vitro transcription and innate immunity activation. Ijms-21-06582-g002.webp
mRNA in vitro transcription and innate immunity activation.

A ribonucleic acid (RNA) vaccine or messenger RNA (mRNA) vaccine is a type of vaccine that uses a copy of a molecule called messenger RNA (mRNA) to produce an immune response. [1] The vaccine transfects molecules of synthetic RNA into immune cells, where the vaccine functions as mRNA, causing the cells to build foreign protein that would normally be produced by a pathogen (such as a virus) or by a cancer cell. These protein molecules stimulate an adaptive immune response that teaches the body to identify and destroy the corresponding pathogen or cancer cells. [1] The mRNA is delivered by a co-formulation of the RNA encapsulated in lipid nanoparticles that protect the RNA strands and help their absorption into the cells. [2] [3]

Contents

Reactogenicity, the tendency of a vaccine to produce adverse reactions, is similar to that of conventional non-RNA vaccines. [4] People susceptible to an autoimmune response may have an adverse reaction to RNA vaccines. [4] The advantages of RNA vaccines over traditional vaccines are ease of design, speed and lower cost of production, the induction of both cellular and humoral immunity, and lack of interaction with the host's genomic DNA. [5] [6] While some RNA vaccines, such as the Pfizer–BioNTech COVID-19 vaccine, have the disadvantage of requiring ultracold storage before distribution, [1] other mRNA vaccines, such as the Moderna, CureVac, and Walvax COVID-19 vaccines, do not have such requirements. [7] [8]

In RNA therapeutics, mRNA vaccines have attracted considerable interest as COVID-19 vaccines. [1] In December 2020, Pfizer–BioNTech and Moderna obtained approval for their mRNA-based COVID-19 vaccines. On 2 December, the UK Medicines and Healthcare products Regulatory Agency (MHRA) became the first medicines regulator to approve an mRNA vaccine, authorizing the Pfizer–BioNTech vaccine for widespread use. [9] [10] [11] On 11 December, the US Food and Drug Administration (FDA) issued an emergency use authorization for the Pfizer–BioNTech vaccine [12] [13] and a week later similarly approved the Moderna vaccine. [14] [15]

History

Early research

Timeline of some key discoveries and advances in the development of mRNA-based drug technology. Ijms-21-06582-g001.webp
Timeline of some key discoveries and advances in the development of mRNA-based drug technology.

The first successful transfection of mRNA packaged within a liposomal nanoparticle into a cell was published in 1989. [16] [17] "Naked" (or unprotected) mRNA was injected a year later into the muscle of mice. [3] [18] These studies were the first evidence that in vitro transcribed mRNA could deliver the genetic information to produce proteins within living cell tissue [3] and led to the concept proposal of messenger RNA vaccines. [19] [20]

Liposome-encapsulated mRNA was shown in 1993 to stimulate T-cells in mice, [21] [22] and mRNA proved useful two years later to elicit both humoral and cellular immune response against a pathogen. [3] [23] [24]

Development

Successful application of modified nucleosides as a medium to transport mRNA inside cells without setting off the body's defense system was reported in 2005. [3] [25] BioNTech in 2008 and Moderna in 2010 were founded to develop mRNA biotechnologies. [26] [27]

In 2010, DARPA launched ADEPT, a biotechnology research program to develop emerging technologies for the US military. [28] DARPA recognized a year later the potential of nucleic acid technology for defense against pandemics and began to invest in the field through ADEPT. [28] [29] DARPA grants were seen as a vote of confidence that in turn encouraged other government agencies and private investors to invest in mRNA technology. [29] In 2013, DARPA awarded a $25 million grant to Moderna. [30]

mRNA drugs for cardiovascular, metabolic, and renal diseases, as well as selected targets for cancer, were initially linked to serious side effects. [31] [32] mRNA vaccines for human use have been studied for rabies, Zika virus disease, cytomegalovirus, and influenza. [33]

Acceleration

In December 2020, BioNTech and Moderna obtained approval for their mRNA-based COVID-19 vaccines. On 2 December, seven days after its final eight-week trial, the UK Medicines and Healthcare products Regulatory Agency became the first global medicines regulator in history to approve an mRNA vaccine, granting emergency authorization for Pfizer–BioNTech's BNT162b2 COVID-19 vaccine for widespread use. [9] [10] [34] On 11 December, the FDA gave emergency use authorization for the Pfizer–BioNTech COVID-19 vaccine and a week later similar approval for the Moderna COVID-19 vaccine. [35]

Mechanism

An illustration of the mechanism of action of the RNA vaccine RNA vaccine-en.svg
An illustration of the mechanism of action of the RNA vaccine
Video showing how vaccination with an mRNA vaccine works

The goal of a vaccine is to stimulate the adaptive immune system to create antibodies that precisely target that particular pathogen. The markers on the pathogen that the antibodies target are called antigens. [36]

Traditional vaccines stimulate an antibody response by injecting either antigens, an attenuated (weakened) virus, an inactivated (dead) virus, or a recombinant antigen-encoding viral vector (harmless carrier virus with an antigen transgene) into the body. These antigens and viruses are prepared and grown outside the body. [37] [38]

In contrast, mRNA vaccines introduce a short-lived [39] synthetically created fragment of the RNA sequence of a virus into the vaccinated individual. These mRNA fragments are taken up by dendritic cells through phagocytosis. [40] The dendritic cells use their internal machinery (ribosomes) to read the mRNA and produce the viral antigens that the mRNA encodes. [4] The body degrades the mRNA fragments within a few days of introduction. [41] Although non-immune cells can potentially also absorb vaccine mRNA, produce antigens, and display the antigens on their surfaces, dendritic cells absorb the mRNA globules much more readily. [42] The mRNA fragments are translated in the cytoplasm and do not affect the body's somatic DNA, located separately in the cell nucleus. [1] [43] [1]

Once the viral antigens are produced by the host cell, the normal adaptive immune system processes are followed. Antigens are broken down by proteasomes. Class I and class II MHC molecules then attach to the antigen and transport it to the cellular membrane, "activating" the dendritic cell. [44] Once activated, dendritic cells migrate to lymph nodes, where they present the antigen to T cells and B cells. [45] This triggers the production of antibodies specifically targeted to the antigen, ultimately resulting in immunity. [36]

mRNA

mRNA components important for expressing the antigen sequence 41541 2020 159 Fig2 HTML.webp
mRNA components important for expressing the antigen sequence

The central component of a mRNA vaccine is its mRNA construct. [46] The in vitro transcribed mRNA is generated from an engineered plasmid DNA, which has an RNA polymerase promoter and sequence which corresponds to the mRNA construct. By combining T7 phage RNA polymerase and the plasmid DNA, the mRNA can be transcribed in the lab. Efficacy of the vaccine is dependent on the stability and structure of the mRNA construct. [47]

The in vitro transcribed mRNA has the same structural components as natural mRNA in eukaryotic cells. It has a 5' cap, a 5'-untranslated region (UTR) and 3'-UTR, an open reading frame (ORF), which encodes the relevant antigen, and a 3'-poly(A) tail. By modifying these different components of the synthetic mRNA, the stability and translational ability of the mRNA can be enhanced, and in turn, the efficacy of the vaccine improved. [46]

The mRNA can be improved by using synthetic 5'-cap analogues which enhance the stability and increase protein translation. Similarly, regulatory elements in the 5'-untranslated region and the 3'-untranslated region can be altered, and the length of the poly(A) tail optimized, to stabilize the mRNA and increase protein production. The mRNA nucleotides can be modified to both decrease innate immune activation and increase the mRNA's half-life in the host cell. The nucleic acid sequence and codon usage impacts protein translation. Enriching the sequence with guanine-cytosine content improves mRNA stability and half-life and, in turn, protein production. Replacing rare codons with synonymous codons frequently used by the host cell further enhances protein production. [47]

Delivery

Major delivery methods and carrier molecules for mRNA vaccines 12943 2021 1311 Fig3 HTML.webp
Major delivery methods and carrier molecules for mRNA vaccines

For a vaccine to be successful, sufficient mRNA must enter the host cell cytoplasm to stimulate production of the specific antigens. Entry of mRNA molecules, however, faces a number of difficulties. Not only are mRNA molecules too large to cross the cell membrane by simple diffusion, they are also negatively charged like the cell membrane, which causes a mutual electrostatic repulsion. Additionally, mRNA is easily degraded by RNAases in skin and blood.[ citation needed ]

Various methods have been developed to overcome these delivery hurdles. The method of vaccine delivery can be broadly classified by whether mRNA transfer into cells occurs within ( in vivo ) or outside ( ex vivo ) the organism. [48] [3]

Ex vivo

Dendritic cells display antigens on their surfaces, leading to interactions with T cells to initiate an immune response. Dendritic cells can be collected from patients and programmed with the desired mRNA, then administered back into patients to create an immune response. [49]

The simplest way that ex vivo dendritic cells take up mRNA molecules is through endocytosis, a fairly inefficient pathway in the laboratory setting that can be significantly improved through electroporation. [48]

In vivo

Since the discovery that the direct administration of in vitro transcribed mRNA leads to the expression of antigens in the body, in vivo approaches have been investigated. [18] They offer some advantages over ex vivo methods, particularly by avoiding the cost of harvesting and adapting dendritic cells from patients and by imitating a regular infection. [48]

Different routes of injection, such as into the skin, blood, or muscles, result in varying levels of mRNA uptake, making the choice of administration route a critical aspect of in vivo delivery. One study showed, in comparing different routes, that lymph node injection leads to the largest T-cell response. [50]

Naked mRNA injection

Naked mRNA injection means that the delivery of the vaccine is only done in a buffer solution. [51] This mode of mRNA uptake has been known since the 1990s. [18] The first worldwide clinical studies used intradermal injections of naked mRNA for vaccination. [52] [53] A variety of methods have been used to deliver naked mRNA, such as subcutaneous, intravenous, and intratumoral injections. Although naked mRNA delivery causes an immune response, the effect is relatively weak, and after injection the mRNA is often rapidly degraded. [48]

Polymer and peptide vectors

Cationic polymers can be mixed with mRNA to generate protective coatings called polyplexes. These protect the recombinant mRNA from ribonucleases and assist its penetration in cells. Protamine is a natural cationic peptide and has been used to encapsulate mRNA for vaccination. [54] [55]

Lipid nanoparticle vector

Assembly of RNA lipid nanoparticle Vaccines-09-00065-g002.webp
Assembly of RNA lipid nanoparticle

The first time the FDA approved the use of lipid nanoparticles as a drug delivery system was in 2018, when the agency approved the first siRNA drug, Onpattro. [56] Encapsulating the mRNA molecule in lipid nanoparticles was a critical breakthrough for producing viable mRNA vaccines, solving a number of key technical barriers in delivering the mRNA molecule into the host cell. [56] [57] Research into using lipids to deliver siRNA to cells became a foundation for similar research into using lipids to deliver mRNA. [58] However, new lipids had to be invented to encapsulate mRNA strands, which are much longer than siRNA strands. [58]

Principally, the lipid provides a layer of protection against degradation, allowing more robust translational output. In addition, the customization of the lipid's outer layer allows the targeting of desired cell types through ligand interactions. However, many studies have also highlighted the difficulty of studying this type of delivery, demonstrating that there is an inconsistency between in vivo and in vitro applications of nanoparticles in terms of cellular intake. [59] The nanoparticles can be administered to the body and transported via multiple routes, such as intravenously or through the lymphatic system. [56]

One issue with lipid nanoparticles is that several of the breakthroughs leading to the practical use of that technology involve the use of microfluidics. Microfluidic reaction chambers are difficult to scale up, since the entire point of microfluidics is to exploit the microscale behaviors of liquids. The only way around this obstacle is to run an extensive number of microfluidic reaction chambers in parallel, a novel task requiring custom-built equipment. [60] [61] For COVID-19 mRNA vaccines, this was the main manufacturing bottleneck. Pfizer used such a parallel approach to solve the scaling problem. After verifying that impingement jet mixers could not be directly scaled up, [62] Pfizer made about 100 of the little mixers (each about the size of a U.S. half-dollar coin), connected them together with pumps and filters with a "maze of piping," [63] [64] and set up a computer system to regulate flow and pressure through the mixers. [62]

Another issue, with the large-scale use of this delivery method, is the availability of the novel lipids used to create lipid nanoparticles, especially ionizable cationic lipids. Before 2020, such lipids were manufactured in small quantities measured in grams or kilograms, and they were used for medical research and a handful of drugs for rare conditions. As the safety and efficacy of RNA vaccines became clear by late 2020, the few companies able to manufacture the requisite lipids were confronted with the challenge of scaling up production to respond to orders for several tons of lipids. [61] [65]

Viral vector

In addition to non-viral delivery methods, RNA viruses have been engineered to achieve similar immunological responses. Typical RNA viruses used as vectors include retroviruses, lentiviruses, alphaviruses and rhabdoviruses, each of which can differ in structure and function. [66] Clinical studies have utilized such viruses on a range of diseases in model animals such as mice, chicken and primates. [67] [68] [69]

Advantages

Traditional vaccines

Advantages and disadvantages of different types of vaccine platforms Cei13517-fig-0002-m.webp
Advantages and disadvantages of different types of vaccine platforms

RNA vaccines offer specific advantages over traditional vaccines. [5] [4] Because RNA vaccines are not constructed from an active pathogen (or even an inactivated pathogen), they are non-infectious. In contrast, traditional vaccines require the production of pathogens, which, if done at high volumes, could increase the risks of localized outbreaks of the virus at the production facility. [5] Another biological advantage of RNA vaccines is that since the antigens are produced inside the cell, they stimulate cellular immunity, as well as humoral immunity. [6] [70]

RNA vaccines have the production advantage that they can be designed swiftly. Moderna designed their mRNA-1273 vaccine for COVID-19 in 2 days. [71] They can also be manufactured faster, more cheaply, and in a more standardized fashion (with fewer error rates in production), which can improve responsiveness to serious outbreaks. [4] [5]

The Pfizer–BioNTech vaccine originally required 110 days to mass produce (before Pfizer began to optimize the manufacturing process to only 60 days), which was substantially faster than traditional flu and polio vaccines. [63] Within that larger timeframe, the actual production time is only about 22 days: two weeks for molecular cloning of DNA plasmids and purification of DNA, four days for DNA-to-RNA transcription and purification of mRNA, and four days to encapsulate mRNA in lipid nanoparticles followed by fill and finish. [72] The majority of the days needed for each production run are allocated to rigorous quality control at each stage. [63]

DNA vaccines

In addition to sharing the advantages of theoretical DNA vaccines over established traditional vaccines, RNA vaccines also have additional advantages over DNA vaccines. The mRNA is translated in the cytosol, so there is no need for the RNA to enter the cell nucleus, and the risk of being integrated into the host genome is averted. [3] Modified nucleosides (for example, pseudouridines, 2'-O-methylated nucleosides) can be incorporated to mRNA to suppress immune response stimulation to avoid immediate degradation and produce a more persistent effect through enhanced translation capacity. [73] [74] [75] The open reading frame (ORF) and untranslated regions (UTR) of mRNA can be optimized for different purposes (a process called sequence engineering of mRNA), for example through enriching the guanine-cytosine content or choosing specific UTRs known to increase translation. [40] An additional ORF coding for a replication mechanism can be added to amplify antigen translation and therefore immune response, decreasing the amount of starting material needed. [76] [77]

Disadvantages

Side effects

Reactogenicity is similar to that of conventional, non-RNA vaccines. However, those susceptible to an autoimmune response may have an adverse reaction to RNA vaccines. [4] The mRNA strands in the vaccine may elicit an unintended immune reaction this entails the body believing itself to be sick, and the person feeling as if they are as a result. To minimize this, mRNA sequences in mRNA vaccines are designed to mimic those produced by host cells. [5]

Strong but transient reactogenic effects were reported in trials of novel COVID-19 RNA vaccines; most people will not experience severe side effects which include fever and fatigue. Severe side effects are defined as those that prevent daily activity. [78]

Recent

Before 2020, no mRNA technology platform (drug or vaccine) had been authorized for use in humans, so there was a risk of unknown effects. [70] The 2020 COVID-19 pandemic required faster production capability of mRNA vaccines, made them attractive to national health organisations, and led to debate about the type of initial authorization mRNA vaccines should get (including emergency use authorization or expanded access authorization) after the eight-week period of post-final human trials. [79] [80]

Storage

Because mRNA is fragile, some vaccines must be kept at very low temperatures to avoid degrading and thus giving little effective immunity to the recipient. Pfizer–BioNTech's BNT162b2 mRNA vaccine has to be kept between −80 and −60 °C (−112 and −76 °F). [81] [82] Moderna says their mRNA-1273 vaccine can be stored between −25 and −15 °C (−13 and 5 °F), [83] which is comparable to a home freezer, [82] and that it remains stable between 2 and 8 °C (36 and 46 °F) for up to 30 days. [83] [84] In November 2020, Nature reported, "While it's possible that differences in LNP formulations or mRNA secondary structures could account for the thermostability differences [between Moderna and BioNtech], many experts suspect both vaccine products will ultimately prove to have similar storage requirements and shelf lives under various temperature conditions." [70] Several platforms are being studied that may allow storage at higher temperatures. [4]

Efficacy

The COVID-19 mRNA vaccines from Moderna and Pfizer–BioNTech have efficacy rates of 90 to 95 percent. Prior mRNA drug trials on pathogens other than COVID-19 were not effective and had to be abandoned in the early phases of trials. The reason for the efficacy of the new mRNA vaccines is not clear. [85]

Physician-scientist Margaret Liu stated that the efficacy of the new COVID-19 mRNA vaccines could be due to the "sheer volume of resources" that went into development, or that the vaccines might be "triggering a nonspecific inflammatory response to the mRNA that could be heightening its specific immune response, given that the modified nucleoside technique reduced inflammation but hasn't eliminated it completely", and that "this may also explain the intense reactions such as aches and fevers reported in some recipients of the mRNA SARS-CoV-2 vaccines". These reactions though severe were transient and another view is that they were believed to be a reaction to the lipid drug delivery molecules. [85]

Hesitancy

There is misinformation implying that mRNA vaccines could alter DNA in the nucleus. [86] mRNA in the cytosol is very rapidly degraded before it would have time to gain entry into the cell nucleus. (mRNA vaccines must be stored at very low temperature to prevent mRNA degradation.) Retrovirus can be single-stranded RNA (just as SARS-CoV-2 vaccine is single-stranded RNA) which enters the cell nucleus and uses reverse transcriptase to make DNA from the RNA in the cell nucleus. A retrovirus has mechanisms to be imported into the nucleus, but other mRNA lack these mechanisms. Once inside the nucleus, creation of DNA from RNA cannot occur without a primer, which accompanies a retrovirus, but which would not exist for other mRNA if placed in the nucleus. [87]

Categories

Non-amplifying mRNA

Mechanism of non-amplifying and self-amplifiying mRNA vaccines 41541 2020 159 Fig1 HTML.webp
Mechanism of non-amplifying and self-amplifiying mRNA vaccines

The two main categories of mRNA vaccines are non-amplifying (conventional) mRNA and molecular self-amplifiying mRNA. [88] [89] Self-amplifying mRNA (saRNA) vaccines began development in the 1990s. [90] [76]

Self-amplifying mRNA

Self-amplifying mRNA is a technology similar to conventional mRNA, except the saRNA produces multiple copies of itself in the cell before producing proteins like mRNA does. [88] [89] This allows smaller quantities to be used and has other potential advantages. [91] [92] The mechanisms and consequently the evaluation of self-amplifying mRNA may be different, as self-amplifying mRNA is fundamentally different by being a much bigger molecule in size. [3]

saRNA vaccines are being researched, including development of a malaria vaccine. [93] In September 2021, Gritstone bio, Inc. started a Phase 1 trial of an saRNA COVID-19 vaccine, used as a booster vaccine. The vaccine is designed to target both the spike protein of the SARS‑CoV‑2 virus, and viral proteins that may be less prone to genetic variation, to provide greater protection against SARS‑CoV‑2 variants. [94] [95]

See also

Related Research Articles

DNA vaccine Vaccine containing DNA

A DNA vaccine is a type of vaccine that transfects a specific antigen-coding DNA sequence into the cells of an organism as a mechanism to induce an immune response.

PEGylation Chemical reaction

PEGylation is the process of both covalent and non-covalent attachment or amalgamation of polyethylene glycol polymer chains to molecules and macrostructures, such as a drug, therapeutic protein or vesicle, which is then described as PEGylated.

Solid lipid nanoparticle Novel drug delivery system

Solid lipid nanoparticles, or lipid nanoparticles (LNPs), are nanoparticles composed of lipids. They are a novel pharmaceutical drug delivery system, and a novel pharmaceutical formulation. LNPs as a drug delivery vehicle were first approved in 2018 for the siRNA drug Onpattro. LNPs became more widely known in late 2020, as some COVID-19 vaccines that use RNA vaccine technology coat the fragile mRNA strands with PEGylated lipid nanoparticles as their delivery vehicle. .

Moderna American biotechnology company innovating on mRNA-based medicines

Moderna, Inc is an American pharmaceutical and biotechnology company based in Cambridge, Massachusetts. It focuses on vaccine technologies based on messenger RNA (mRNA). Moderna's vaccine platform inserts synthetic nucleoside-modified messenger RNA (modRNA) into human cells using a coating of lipid nanoparticles. This mRNA then reprograms the cells to prompt immune responses. Moderna develops mRNA therapeutic vaccines that are delivered in lipid nanoparticles, using mRNA with pseudouridine nucleosides. Candidates are designed to have improved folding and translation efficiency via insertional mutagenesis.

Arcturus Therapeutics is an American RNA medicines biotechnology company focused on the discovery, development and commercialization of therapeutics for rare diseases and infectious diseases. Arcturus has developed a novel, potent, and safe RNA therapeutics platform called LUNAR, a proprietary lipid-enabled delivery system for nucleic acid medicines including small interfering RNA (siRNA), messenger RNA (mRNA), gene editing RNA, DNA, antisense oligonucleotides (ASO), and microRNA.

Julianna Lisziewicz is a Hungarian immunologist. Lisziewicz headed many research teams that have discovered and produced immunotheraputic drugs to treat diseases like cancer and chronic infections like HIV/AIDS. Some of these drugs have been successfully used in clinical trials.

COVID-19 vaccine Vaccine designed to provide acquired immunity against SARS-CoV-2

A COVID‑19 vaccine is a vaccine intended to provide acquired immunity against severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2), the virus that causes coronavirus disease 2019 (COVID‑19). Prior to the COVID‑19 pandemic, an established body of knowledge existed about the structure and function of coronaviruses causing diseases like severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS). This knowledge accelerated the development of various vaccine platforms during early 2020. The initial focus of SARS-CoV-2 vaccines was on preventing symptomatic, often severe illness. On 10 January 2020, the SARS-CoV-2 genetic sequence data was shared through GISAID, and by 19 March, the global pharmaceutical industry announced a major commitment to address COVID-19. The COVID‑19 vaccines are widely credited for their role in reducing the spread, severity, and death caused by COVID-19.

Moderna COVID-19 vaccine RNA COVID-19 vaccine

The Moderna COVID‑19 vaccine, codenamed mRNA-1273 and sold under the brand name Spikevax, is a COVID-19 vaccine developed by American company Moderna, the United States National Institute of Allergy and Infectious Diseases (NIAID) and the Biomedical Advanced Research and Development Authority (BARDA). It is authorized for use in people aged twelve years and older in some jurisdictions and for people eighteen years and older in other jurisdictions to provide protection against COVID-19 which is caused by infection by the SARS-CoV-2 virus. It is designed to be administered as two or three 0.5 mL doses given by intramuscular injection at an interval of at least 28 days apart.

Katalin Karikó Hungarian biochemist (born 1955)

Katalin Karikó is a Hungarian biochemist who specializes in RNA-mediated mechanisms. Her research has been the development of in vitro-transcribed mRNA for protein therapies. She co-founded and was CEO of RNARx, from 2006 to 2013. Since 2013, she has been associated with BioNTech RNA Pharmaceuticals, first as a vice president and promoted to senior vice president in 2019. She also is an adjunct professor at the University of Pennsylvania.

BioNTech German biotechnology company

BioNTech SE is a German biotechnology company based in Mainz that develops and manufactures active immunotherapies for patient-specific approaches to the treatment of diseases. It develops pharmaceutical candidates based on messenger ribonucleic acid (mRNA) for use as individualized cancer immunotherapies, as vaccines against infectious diseases and as protein replacement therapies for rare diseases, and also engineered cell therapy, novel antibodies and small molecule immunomodulators as treatment options for cancer.

RNA therapeutics are a new class of medications based on ribonucleic acid (RNA). Research has been working on clinical use since the 1990s, with significant success in cancer therapy in the early 2010s. In 2020 and 2021, mRNA vaccines have been developed globally for use in combating the coronavirus disease. The Pfizer–BioNTech COVID-19 vaccine was the first mRNA vaccine approved by a medicines regulator, followed by the Moderna COVID-19 vaccine, and others.

Uğur Şahin German oncologist

Uğur Şahin is a German oncologist, immunologist and CEO of BioNTech, which helped develop one of the major vaccines against COVID-19. His main fields of research are cancer research and immunology.

Pfizer–BioNTech COVID-19 vaccine mRNA-based COVID-19 vaccine

The Pfizer–BioNTech COVID-19 vaccine, sold under the brand name Comirnaty, is an mRNA-based COVID-19 vaccine developed by the German biotechnology company BioNTech and for its development collaborated with American company Pfizer, for support with clinical trials, logistics, and manufacturing. It is authorized for use in people aged twelve years and older in some jurisdictions and for people sixteen years and older in other jurisdictions, to provide protection against COVID-19, caused by infection with the SARS-CoV-2 virus. The vaccine is given by intramuscular injection. It is composed of nucleoside-modified mRNA (modRNA) encoding a mutated form of the full-length spike protein of SARS-CoV-2, which is encapsulated in lipid nanoparticles. Initial advice indicated that vaccination required two doses given 21 days apart, but the interval was later extended to up to 42 days in the US, and up to four months in Canada.

A nucleoside-modified messenger RNA (modRNA) is a synthetic messenger RNA (mRNA) in which some nucleosides are replaced by other naturally modified nucleosides or by synthetic nucleoside analogues. modRNA is used to induce the production of a desired protein in certain cells. An important application is the development of mRNA vaccines, of which the first authorized were COVID-19 vaccines.

ALC-0315 Chemical compound

ALC-0315 is a synthetic lipid. A colorless oily material, it has attracted attention as a component of the SARS-CoV-2 vaccine, BNT162b2, from BioNTech and Pfizer. Specifically, it is one of four components that form lipid nanoparticles (LNPs), which encapsulate and protect the otherwise fragile mRNA that is the active ingredient in these drugs. These nanoparticles promote the uptake of therapeutically effective nucleic acids such as oligonucleotides or mRNA both in vitro and in vivo.

Distearoylphosphatidylcholine(DSPC) is a phosphatidylcholine, a kind of phospholipid. It is a natural constituent of cell membranes, eg. soybean phosphatidylcholines are mostly different 18-carbon phosphatidylcholines, and their hydrogenation results in 85% DSPC. It can be used to prepare lipid nanoparticles which are used in mRNA vaccines, In particular, it forms part of the drug delivery system for the Moderna and Pfizer COVID-19 vaccines.

Drew Weissman American medical academic

Drew Weissman is a physician-scientist best known for his contributions to RNA biology. His work helped enable development of effective mRNA vaccines, the best known of which are those for COVID-19 produced by BioNTech/Pfizer and Moderna. Weissman is a professor of medicine at the Perelman School of Medicine at the University of Pennsylvania (Penn).

SM-102 is a synthetic amino lipid which is used in combination with other lipids to form lipid nanoparticles. These are used for the delivery of mRNA-based vaccines, and in particular SM-102 forms part of the drug delivery system for the Moderna COVID-19 vaccine.

CureVac COVID-19 vaccine Vaccine candidate against COVID-19

The CureVac COVID-19 vaccine is a COVID-19 vaccine candidate developed by CureVac N.V. and the Coalition for Epidemic Preparedness Innovations (CEPI). The vaccine showed inadequate results in its Phase III trials with only 47% efficacy.

COVID-19 vaccine clinical research

COVID-19 vaccine clinical research is the clinical research on COVID-19 vaccines, including their efficacy, effectiveness and safety. There are 22 vaccines authorized for use by national governments, with six vaccines being approved for emergency or full use by at least one WHO-recognised stringent regulatory authority; and five of them are in Phase IV. 204 vaccines under clinical trials that have not yet been authorized. There are also nine clinical trials on heterologous vaccination courses.

References

  1. 1 2 3 4 5 6 Park KS, Sun X, Aikins ME, Moon JJ (December 2020). "Non-viral COVID-19 vaccine delivery systems". Advanced Drug Delivery Reviews. 169: 137–51. doi:10.1016/j.addr.2020.12.008. PMC   7744276 . PMID   33340620.
  2. Kowalski PS, Rudra A, Miao L, Anderson DG (April 2019). "Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery". Mol Ther. 27 (4): 710–28. doi:10.1016/j.ymthe.2019.02.012. PMC   6453548 . PMID   30846391.
  3. 1 2 3 4 5 6 7 8 Verbeke R, Lentacker I, De Smedt SC, Dewitte H (October 2019). "Three decades of messenger RNA vaccine development". Nano Today. 28: 100766. doi:10.1016/j.nantod.2019.100766. hdl: 1854/LU-8628303 .
  4. 1 2 3 4 5 6 7 Pardi N, Hogan MJ, Porter FW, Weissman D (April 2018). "mRNA vaccines – a new era in vaccinology". Nature Reviews. Drug Discovery. 17 (4): 261–79. doi:10.1038/nrd.2017.243. PMC   5906799 . PMID   29326426.
  5. 1 2 3 4 5 PHG Foundation (2019). "RNA vaccines: an introduction". University of Cambridge . Retrieved 18 November 2020.
  6. 1 2 Kramps T, Elders K (2017). "Introduction to RNA Vaccines". RNA Vaccines: Methods and Protocols. Methods in Molecular Biology. 1499. pp. 1–11. doi:10.1007/978-1-4939-6481-9_1. ISBN   978-1-4939-6479-6. PMID   27987140.
  7. Crommelin DJ, Anchordoquy TJ, Volkin DB, Jiskoot W, Mastrobattista E (March 2021). "Addressing the Cold Reality of mRNA Vaccine Stability". Journal of Pharmaceutical Sciences. 110 (3): 997–1001. doi:10.1016/j.xphs.2020.12.006. PMC   7834447 . PMID   33321139.
  8. "Mexico to start late-stage clinical trial for China's mRNA COVID-19 vaccine". Reuters. 11 May 2021. Retrieved 19 August 2021.
  9. 1 2 "UK authorises Pfizer/BioNTech COVID-19 vaccine" (Press release). Department of Health and Social Care. 2 December 2020.
  10. 1 2 Boseley S, Halliday J (2 December 2020). "UK approves Pfizer/BioNTech Covid vaccine for rollout next week". The Guardian . Retrieved 2 December 2020.
  11. "Conditions of Authorisation for Pfizer/BioNTech COVID-19 Vaccine" (Decision). Medicines & Healthcare Products Regulatory Agency. 8 December 2020.
  12. "FDA Takes Key Action in Fight Against COVID-19 By Issuing Emergency Use Authorization for First COVID-19 Vaccine". U.S. Food and Drug Administration (FDA) (Press release). 11 December 2020. Retrieved 6 February 2021.
  13. Oliver SE, Gargano JW, Marin M, Wallace M, Curran KG, Chamberland M, et al. (December 2020). "The Advisory Committee on Immunization Practices' Interim Recommendation for Use of Pfizer-BioNTech COVID-19 Vaccine – United States, December 2020" (PDF). MMWR Morb Mortal Wkly Rep. 69 (50): 1922–24. doi:10.15585/mmwr.mm6950e2. PMC   7745957 . PMID   33332292.
  14. "FDA Takes Additional Action in Fight Against COVID-19 By Issuing Emergency Use Authorization for Second COVID-19 Vaccine". U.S. Food and Drug Administration (FDA) (Press release). 18 December 2020.
  15. Oliver SE, Gargano JW, Marin M, Wallace M, Curran KG, Chamberland M, et al. (January 2021). "The Advisory Committee on Immunization Practices' Interim Recommendation for Use of Moderna COVID-19 Vaccine – United States, December 2020" (PDF). MMWR Morb Mortal Wkly Rep. 69 (5152): 1653–56. doi: 10.15585/mmwr.mm695152e1 . PMID   33382675. S2CID   229945697.
  16. Xu S, Yang K, Li R, Zhang L (September 2020). "mRNA Vaccine Era-Mechanisms, Drug Platform and Clinical Prospection". International Journal of Molecular Sciences. 21 (18): 6582. doi: 10.3390/ijms21186582 . PMC   7554980 . PMID   32916818. Initiation of cationic lipid-mediated mrna transfection; Concept proposal of mRNA-based drugs.
  17. Malone RW, Felgner PL, Verma IM (August 1989). "Cationic liposome-mediated RNA transfection". Proceedings of the National Academy of Sciences of the United States of America. 86 (16): 6077–81. Bibcode:1989PNAS...86.6077M. doi: 10.1073/pnas.86.16.6077 . PMC   297778 . PMID   2762315.
  18. 1 2 3 Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL (March 1990). "Direct gene transfer into mouse muscle in vivo". Science. 247 (4949 Pt 1): 1465–8. Bibcode:1990Sci...247.1465W. doi:10.1126/science.1690918. PMID   1690918.
  19. May M (31 May 2021). "After COVID-19 successes, researchers push to develop mRNA vaccines for other diseases". Nature . Retrieved 31 July 2021. When the broad range of vaccines against COVID-19 were being tested in clinical trials, only a few experts expected the unproven technology of mRNA to be the star. Within 10 months, mRNA vaccines were both the first to be approved and the most effective. Although these are the first mRNA vaccines to be approved, the story of mRNA vaccines starts more than 30 years ago, with many bumps in the road along the way. In 1990, the late physician-scientist Jon Wolff and his University of Wisconsin colleagues injected mRNA into mice, which caused cells in the mice to produce the encoded proteins. In many ways, that work served as the first step toward making a vaccine from mRNA, but there was a long way to go—and there still is, for many applications.
  20. Xu S, Yang K, Li R, Zhang L (September 2020). "mRNA Vaccine Era-Mechanisms, Drug Platform and Clinical Prospection". International Journal of Molecular Sciences. 21 (18): 6582. doi: 10.3390/ijms21186582 . PMC   7554980 . PMID   32916818. Concept proposal of mRNA vaccines (1990)
  21. Pascolo S (August 2004). "Messenger RNA-based vaccines". Expert Opinion on Biological Therapy. 4 (8): 1285–94. doi:10.1517/14712598.4.8.1285. PMID   15268662. S2CID   19350848.
  22. Martinon F, Krishnan S, Lenzen G, Magné R, Gomard E, Guillet JG, et al. (July 1993). "Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA". European Journal of Immunology. 23 (7): 1719–22. doi:10.1002/eji.1830230749. PMID   8325342. S2CID   42640967.
  23. Kallen KJ, Theß A (January 2014). "A development that may evolve into a revolution in medicine: mRNA as the basis for novel, nucleotide-based vaccines and drugs". Therapeutic Advances in Vaccines. 2 (1): 10–31. doi:10.1177/2051013613508729. PMC   3991152 . PMID   24757523.
  24. Conry RM, LoBuglio AF, Wright M, Sumerel L, Pike MJ, Johanning F, et al. (April 1995). "Characterization of a messenger RNA polynucleotide vaccine vector". Cancer Research. 55 (7): 1397–400. PMID   7882341.
  25. Karikó K, Buckstein M, Ni H, Weissman D (August 2005). "Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA". Immunity. 23 (2): 165–75. doi: 10.1016/j.immuni.2005.06.008 . PMID   16111635.
  26. "BioNTech's founders: scientist couple in global spotlight". France 24. 13 November 2020. Retrieved 31 July 2021.
  27. Garade D (10 November 2020). "The story of mRNA: How a once-dismissed idea became a leading technology in the Covid vaccine race". Stat . Retrieved 16 November 2020.
  28. 1 2 Sonne P (30 July 2020). "How a secretive Pentagon agency seeded the ground for a rapid coronavirus cure". The Washington Post.
  29. 1 2 Usdin S (19 March 2020). "DARPA's gambles might have created the best hopes for stopping COVID-19". BioCentury. Retrieved 19 June 2021.
  30. "DARPA Awards Moderna Therapeutics A Grant For Up To $25 Million To Develop Messenger RNA Therapeutics". 2 October 2013. Retrieved 31 May 2021.
  31. Garde D (10 January 2017). "Lavishly funded Moderna hits safety problems in bold bid to revolutionize medicine". Stat . Archived from the original on 16 November 2020. Retrieved 19 May 2020. struggling to get mRNA into cells without triggering nasty side effects
  32. Garade D (13 September 2016). "Ego, ambition, and turmoil: Inside one of biotech's most secretive startups". Stat . Archived from the original on 16 November 2020. Retrieved 18 May 2020. because it’s exceedingly hard to get RNA into cells without triggering nasty side effects
  33. "COVID-19 and Your Health". Centers for Disease Control and Prevention. 11 February 2020.
  34. Roberts M (2 December 2020). "Covid Pfizer vaccine approved for use next week in UK". BBC News. Retrieved 2 December 2020.
  35. Office of the Commissioner (18 December 2020). "Pfizer-BioNTech COVID-19 Vaccine". FDA.
  36. 1 2 Batty CJ, Heise MT, Bachelder EM, Ainslie KM (December 2020). "Vaccine formulations in clinical development for the prevention of severe acute respiratory syndrome coronavirus 2 infection". Advanced Drug Delivery Reviews. 169: 168–89. doi:10.1016/j.addr.2020.12.006. PMC   7733686 . PMID   33316346.
  37. Kyriakidis NC, López-Cortés A, González EV, Grimaldos AB, Prado EO (February 2021). "SARS-CoV-2 vaccines strategies: a comprehensive review of phase 3 candidates". NPJ Vaccines. 6 (1): 28. doi:10.1038/s41541-021-00292-w. PMC   7900244 . PMID   33619260.
  38. Bull JJ, Nuismer SL, Antia R (July 2019). "Recombinant vector vaccine evolution". PLOS Computational Biology. 15 (7): e1006857. Bibcode:2019PLSCB..15E6857B. doi:10.1371/journal.pcbi.1006857. PMC   6668849 . PMID   31323032.
  39. Hajj KA, Whitehead KA (12 September 2017). "Tools for translation: non-viral materials for therapeutic mRNA delivery". Nature Reviews Materials. 2 (10): 17056. Bibcode:2017NatRM...217056H. doi: 10.1038/natrevmats.2017.56 .
  40. 1 2 Schlake T, Thess A, Fotin-Mleczek M, Kallen KJ (November 2012). "Developing mRNA-vaccine technologies". RNA Biology. 9 (11): 1319–30. doi:10.4161/rna.22269. PMC   3597572 . PMID   23064118.
  41. Anand P, Stahel VP (May 2021). "Review the safety of Covid-19 mRNA vaccines: a review". Patient Safety in Surgery. 15 (1): 20. doi:10.1186/s13037-021-00291-9. PMC   8087878 . PMID   33933145.
  42. Goldman B (22 December 2020). "How do the new COVID-19 vaccines work?". Scope. Stanford Medicine. Retrieved 28 January 2021.
  43. Xu, Shuqin; Yang, Kunpeng; Li, Rose; Zhang, Lu (9 September 2020). "mRNA Vaccine Era—Mechanisms, Drug Platform and Clinical Prospection". International Journal of Molecular Sciences. 21 (18): 6582. doi:10.3390/ijms21186582. PMC   7554980 .
  44. Xu S, Yang K, Li R, Zhang L (September 2020). "mRNA Vaccine Era-Mechanisms, Drug Platform and Clinical Prospection". International Journal of Molecular Sciences. 21 (18): 6582. doi: 10.3390/ijms21186582 . PMC   7554980 . PMID   32916818.
  45. Fiedler K, Lazzaro S, Lutz J, Rauch S, Heidenreich R (2016). "mRNA Cancer Vaccines". Recent Results in Cancer Research. Fortschritte der Krebsforschung. Progres dans les Recherches Sur le Cancer. Recent Results in Cancer Research. 209: 61–85. doi:10.1007/978-3-319-42934-2_5. ISBN   978-3-319-42932-8. PMID   28101688.
  46. 1 2 Jackson, Nicholas A. C.; Kester, Kent E.; Casimiro, Danilo; Gurunathan, Sanjay; DeRosa, Frank (4 February 2020). "The promise of mRNA vaccines: a biotech and industrial perspective". npj Vaccines. 5 (1): 1–6. doi:10.1038/s41541-020-0159-8. ISSN   2059-0105. PMC   7000814 .
  47. 1 2 Pardi, Norbert; Hogan, Michael J.; Porter, Frederick W.; Weissman, Drew (April 2018). "mRNA vaccines — a new era in vaccinology". Nature Reviews Drug Discovery. 17 (4): 261–279. doi:10.1038/nrd.2017.243. ISSN   1474-1784. PMC   5906799 .
  48. 1 2 3 4 Xu S, Yang K, Li R, Zhang L (September 2020). "mRNA Vaccine Era-Mechanisms, Drug Platform and Clinical Prospection". International Journal of Molecular Sciences. 21 (18): 6582. doi:10.3390/ijms21186582. PMC   7554980 . PMID   32916818.
  49. Benteyn D, Heirman C, Bonehill A, Thielemans K, Breckpot K (February 2015). "mRNA-based dendritic cell vaccines". Expert Review of Vaccines. 14 (2): 161–76. doi:10.1586/14760584.2014.957684. PMID   25196947. S2CID   38292712.
  50. Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, et al. (November 2010). "Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity". Cancer Research. 70 (22): 9031–40. doi: 10.1158/0008-5472.can-10-0699 . PMID   21045153.
  51. "Vaccine components". Immunisation Advisory Centre. 22 September 2016. Retrieved 20 December 2020.
  52. Probst J, Weide B, Scheel B, Pichler BJ, Hoerr I, Rammensee HG, Pascolo S (August 2007). "Spontaneous cellular uptake of exogenous messenger RNA in vivo is nucleic acid-specific, saturable and ion dependent". Gene Therapy. 14 (15): 1175–80. doi: 10.1038/sj.gt.3302964 . PMID   17476302.
  53. Lorenz C, Fotin-Mleczek M, Roth G, Becker C, Dam TC, Verdurmen WP, et al. (July 2011). "Protein expression from exogenous mRNA: uptake by receptor-mediated endocytosis and trafficking via the lysosomal pathway". RNA Biology. 8 (4): 627–36. doi: 10.4161/rna.8.4.15394 . PMID   21654214.
  54. [ non-primary source needed ]Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, et al. (June 2009). "Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients". Journal of Immunotherapy. 32 (5): 498–507. doi:10.1097/CJI.0b013e3181a00068. PMID   19609242. S2CID   3278811.
  55. Wang Y, Zhang Z, Luo J, Han X, Wei Y, Wei X (February 2021). "mRNA vaccine: a potential therapeutic strategy". Molecular Cancer. 20 (1): 33. doi:10.1186/s12943-021-01311-z. PMC   7884263 . PMID   33593376.
  56. 1 2 3 Cooney E (1 December 2020). "How nanotechnology helps mRNA Covid-19 vaccines work". Stat . Retrieved 3 December 2020.
  57. Reichmuth AM, Oberli MA, Jaklenec A, Langer R, Blankschtein D (May 2016). "mRNA vaccine delivery using lipid nanoparticles". Therapeutic Delivery. 7 (5): 319–34. doi:10.4155/tde-2016-0006. PMC   5439223 . PMID   27075952.
  58. 1 2 Cross R (6 March 2021). "Without these lipid shells, there would be no mRNA vaccines for COVID-19". Chemical & Engineering News. American Chemical Society. Retrieved 6 March 2021.
  59. Paunovska K, Sago CD, Monaco CM, Hudson WH, Castro MG, Rudoltz TG, et al. (March 2018). "A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation". Nano Letters. 18 (3): 2148–57. Bibcode:2018NanoL..18.2148P. doi:10.1021/acs.nanolett.8b00432. PMC   6054134 . PMID   29489381.
  60. Lowe D (3 February 2021). "Opinion: A straightforward explanation why more COVID-19 vaccines can't be produced with help from 'dozens' of companies". MarketWatch. Retrieved 5 February 2021.
  61. 1 2 King A (23 March 2021). "Why manufacturing Covid vaccines at scale is hard". Chemistry World. Royal Society of Chemistry. Retrieved 26 March 2021.
  62. 1 2 Sealy A (2 April 2021). "Manufacturing moonshot: How Pfizer makes its millions of Covid-19 vaccine doses". CNN.
  63. 1 2 3 Weise E, Weintraub K (7 February 2021). "Race to the Vaccine: A COVID-19 vaccine life cycle: from DNA to doses". USA Today. Gannett. Retrieved 24 February 2021.
  64. Hopkins JS, Eastwood J, Moriarty D (3 March 2021). "mRNA Covid-19 Vaccines Are Fast to Make, but Hard to Scale". The Wall Street Journal.
  65. Rowland C (18 February 2021). "Why grandparents can't find vaccines: Scarcity of niche biotech ingredients". The Washington Post.
  66. Lundstrom K (March 2019). "RNA Viruses as Tools in Gene Therapy and Vaccine Development". Genes. 10 (3): 189. doi: 10.3390/genes10030189 . PMC   6471356 . PMID   30832256.
  67. Huang TT, Parab S, Burnett R, Diago O, Ostertag D, Hofman FM, et al. (February 2015). "Intravenous administration of retroviral replicating vector, Toca 511, demonstrates therapeutic efficacy in orthotopic immune-competent mouse glioma model". Human Gene Therapy. 26 (2): 82–93. doi:10.1089/hum.2014.100. PMC   4326030 . PMID   25419577.
  68. Schultz-Cherry S, Dybing JK, Davis NL, Williamson C, Suarez DL, Johnston R, Perdue ML (December 2000). "Influenza virus (A/HK/156/97) hemagglutinin expressed by an alphavirus replicon system protects chickens against lethal infection with Hong Kong-origin H5N1 viruses". Virology. 278 (1): 55–59. doi: 10.1006/viro.2000.0635 . PMID   11112481.
  69. Geisbert TW, Feldmann H (November 2011). "Recombinant vesicular stomatitis virus-based vaccines against Ebola and Marburg virus infections". The Journal of Infectious Diseases. 204 (Suppl 3): S1075–81. doi:10.1093/infdis/jir349. PMC   3218670 . PMID   21987744.
  70. 1 2 3 Dolgin E (November 2020). "COVID-19 vaccines poised for launch, but impact on pandemic unclear". Nature Biotechnology. doi:10.1038/d41587-020-00022-y. PMID   33239758. S2CID   227176634.
  71. Neilson S, Dunn A, Bendix A (26 November 2020). "Moderna's groundbreaking coronavirus vaccine was designed in just 2 days". Business Insider . Retrieved 28 November 2020.
  72. Rabson M (27 February 2021). "From science to syringe: COVID-19 vaccines are miracles of science and supply chains". CTV News. Bell Media.
  73. Karikó K, Buckstein M, Ni H, Weissman D (August 2005). "Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA". Immunity. 23 (2): 165–75. doi: 10.1016/j.immuni.2005.06.008 . PMID   16111635.
  74. Karikó K, Muramatsu H, Ludwig J, Weissman D (November 2011). "Generating the optimal mRNA for therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-modified, protein-encoding mRNA". Nucleic Acids Research. 39 (21): e142. doi:10.1093/nar/gkr695. PMC   3241667 . PMID   21890902.
  75. Pardi N, Weissman D (17 December 2016). "Nucleoside Modified mRNA Vaccines for Infectious Diseases". RNA Vaccines. Methods in Molecular Biology. 1499. Springer New York. pp. 109–21. doi:10.1007/978-1-4939-6481-9_6. ISBN   978-1-4939-6479-6. PMID   27987145.
  76. 1 2 Berglund P, Smerdou C, Fleeton MN, Tubulekas I, Liljeström P (June 1998). "Enhancing immune responses using suicidal DNA vaccines". Nature Biotechnology. 16 (6): 562–65. doi:10.1038/nbt0698-562. PMID   9624688. S2CID   38532700.
  77. Vogel AB, Lambert L, Kinnear E, Busse D, Erbar S, Reuter KC, et al. (February 2018). "Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to mRNA Vaccines but at Much Lower Doses". Molecular Therapy. 26 (2): 446–55. doi:10.1016/j.ymthe.2017.11.017. PMC   5835025 . PMID   29275847.
  78. Wadman M (November 2020). "Public needs to prep for vaccine side effects". Science. 370 (6520): 1022. doi: 10.1126/science.370.6520.1022 . PMID   33243869.
  79. Thomas K (22 October 2020). "Experts Tell F.D.A. It Should Gather More Safety Data on Covid-19 Vaccines". New York Times . Retrieved 21 November 2020.
  80. Kuchler H (30 September 2020). "Pfizer boss warns on risk of fast-tracking vaccines". Financial Times . Retrieved 21 November 2020.
  81. "Pfizer-BioNTech COVID-19 Vaccine Vaccination Storage & Dry Ice Safety Handling". Pfizer. Retrieved 17 December 2020.
  82. 1 2 Simmons-Duffin S. "Why Does Pfizer's COVID-19 Vaccine Need To Be Kept Colder Than Antarctica?". NPR.org. Retrieved 18 November 2020.
  83. 1 2 "Fact Sheet for Healthcare Providers Administering Vaccine" (PDF). ModernaTX, Inc.
  84. "Moderna Announces Longer Shelf Life for its COVID-19 Vaccine Candidate at Refrigerated Temperatures". NPR.org.
  85. 1 2 Kwon D (25 November 2020). "The Promise of mRNA Vaccines". The Scientist . Retrieved 27 November 2020.
  86. Carmichael F, Goodman J (2 December 2020). "Vaccine rumours debunked: Microchips, 'altered DNA' and more" (Reality Check). BBC.
  87. Skalka AM (2014). "Retroviral DNA Transposition: Themes and Variations". Microbiology Spectrum . 2 (5): 1101–23. doi:10.1128/microbiolspec.MDNA3-0005-2014. ISBN   9781555819200. PMC   4383315 . PMID   25844274.
  88. 1 2 Deering RP, Kommareddy S, Ulmer JB, Brito LA, Geall AJ (June 2014). "Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines". Expert Opin Drug Deliv. 11 (6): 885–99. doi:10.1517/17425247.2014.901308. PMID   24665982. S2CID   33489182.
  89. 1 2 Geall AJ, Verma A, Otten GR, Shaw CA, Hekele A, Banerjee K, Cu Y, Beard CW, Brito LA, Krucker T, O'Hagan DT, Singh M, Mason PW, Valiante NM, Dormitzer PR, Barnett SW, Rappuoli R, Ulmer JB, Mandl CW (September 2012). "Nonviral delivery of self-amplifying RNA vaccines". Proc Natl Acad Sci U S A. 109 (36): 14604–09. Bibcode:2012PNAS..10914604G. doi: 10.1073/pnas.1209367109 . PMC   3437863 . PMID   22908294.
  90. Zhou X, Berglund P, Rhodes G, Parker SE, Jondal M, Liljeström P (December 1994). "Self-replicating Semliki Forest virus RNA as recombinant vaccine". Vaccine. 12 (16): 1510–14. doi:10.1016/0264-410x(94)90074-4. PMID   7879415.
  91. Bloom K, van den Berg F, Arbuthnot P (April 2021). "Self-amplifying RNA vaccines for infectious diseases". Gene Therapy. 28 (3–4): 117–129. doi:10.1038/s41434-020-00204-y. PMC   7580817 . PMID   33093657.
  92. "saRNA Biology | About Self-Amplifying RNA Genome & How It Works". Chimeron Bio | Transforming RNA Therapy.
  93. Lowe D (1 March 2021). "A Malaria Vaccine Candidate". Science Translational Medicine . Retrieved 7 May 2021.
  94. Knapton, Sarah (20 September 2021). "First 'variant-proof' Covid vaccine starts trials in Manchester - Retired couple Andrew Clarke, 63, and his wife Helen, 64, from Bolton, became the first to receive the mRNA vaccine on Monday". The Daily Telegraph . Retrieved 21 September 2021.
  95. "Gritstone Announces Dosing of First Volunteer in Trial Evaluating Self-Amplifying mRNA as a COVID-19 Vaccine Booster and Immunogenicity Enhancer". PipelineReview. 20 September 2021. Retrieved 21 September 2021.

Further reading