You are on page 1of 38

MRNA VACCINE

BY

TEMIDAYO FAROUK OLAPADE


18/57MB/00835

A SEMINAR REPORT SUBMITTED TO THE DEPARTMENT OF MICROBIOLOGY,


FACULTY OF PURE AND APPLIED SCIENCES, KWARA STATE UNIVERSITY
MALETE, KWARA STATE.
IN PARTIAL FUFILLMENT OF THE REQUIREMENT FOR THE AWARD OF
BACHELOR OF SCIENCE (B S c) DEGREE IN MICRIOBIOLOGY.

MARCH, 2022

1
TABLE OF CONTENT
TITLE OF PAGE PAGES
INTRODUCTION 1
MRNA 2
MECHANISM OF MRNA VACCINE 4
DELIVERY OF MRNA VACCINE 6
CATEGORIES OF MRNA VACCINE 12
ADVANTAGES OF MRNA VACCINE 14
DISADVANTAGES OF MRNA VACCINE 16
EFFICACY OF MRNA VACCINE 19
HESISTANCY OF MRNA VACCINE 19
PFIZER-BIONTECH COVID-19 VACCINE 20
MANUFACTURING OF PFIZER AND BIONTECH VACCINE 22
MEDICAL USES OF MRNA VACCINE 27
ADVERSE EFFECTS OF MRNA VACCINE 27
CONCLUSIONS 29
RECOMMENDATIONS 30
SUMMARY 31
REFERENCE 32

LIST OF FIGURES
FIGURE NO PAGE
Figure 1. mRNA components important for expressing the antigen sequence, (Jackson et al.,
2020)…………………………………………………………………………………….3
Figure 2. An Illustration of the mechansims of action of a mRNA vaccine (Jackson et al.,
2020)…………………………………………………………………………………….5
Figure 3. Major delivery methods and carrier molecules for mRNA vaccine, (Wang et al.,
2020)……………………………………………………………………………………..7
Figure 4. Assembly of RNA lipid nanoparticles, (Buschmann, 2021)…………………...11
Figure 5. Mechanism of non-amplifying and self-amplifying mRNA vaccine, (Tregoning,
2021)………………………………………………………………………………………13

2
Figure 6. Advantages and disadvantage of different types of vaccine platforms, , (Tregoning,
2021)………………………………………………………………………………………18

LIST OF PLATES
PLATE NO PAGE
Plate 1. A vial of the Pfizer-BioNTech Covid-19 vaccine for the U.S. market, (Jackson et al.,
2020)…………………………………………………………………………………….21
Plate 2. Boxes containing the COVID-19 vaccine of the Pfizer factory in Puurs, (Jackson et
al., 2020)…………………………………………………………………………………….24
Plate 3. A Pfizer employee putting dry ice in a box to protect the COVID-19 vaccine during
transport at the Puurs factory, (Jackson et al., 2020)…………………………………….25
Plate 4. Inside view of Pfixer factory in Puurs, (Jackson et al., 2020)………..………….26

3
INTRODUCTION

An mRNA vaccine is a type of vaccine that uses a copy of a molecule called messenger

RNA (mRNA) to produce an immune response (Park et al., 2020). The

vaccine delivers molecules of antigen-encoding mRNA into immune cells, which use the

designed mRNA as a template to build foreign protein that would normally be produced by

a pathogen 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 (Park et al., 2020). 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 (Kowalski et al., 2019).

Reactogenicity, the tendency of a vaccine to produce adverse reactions, is similar to that of

conventional non-RNA vaccines (Pardi et al., 2018). People susceptible to an autoimmune

response may have an adverse reaction to messenger RNA vaccines (Pardi et al., 2018). The

advantages of mRNA 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 genomic DNA (Pardi et al., 2018). While some messenger RNA

vaccines, such as the Pfizer–BioNTech COVID-19 vaccine, have the disadvantage of

requiring ultracold storage before distribution, (Park et al., 2020) other mRNA vaccines, such

as the Moderna, CureVac, and Walvax COVID-19 vaccines, do not have such requirements

(Crommelin et al., 2021).

In RNA therapeutics, messenger RNA vaccines have attracted considerable interest

as COVID-19 vaccines (Park et al., 2020).  In December 2020, Pfizer–

BioNTech and Moderna obtained authorization 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–

4
BioNTech vaccine for widespread use (Boseley and Halliday, 2020). On 11 December, the

US Food and Drug Administration (FDA) issued an emergency use authorization for the

Pfizer–BioNTech vaccine (FDA, 2020) and a week later similarly authorized the Moderna

vaccine.

MRNA

The central component of a mRNA vaccine is its mRNA construct (Jackson et al.,

2020). 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 designed mRNA (Pardi et al., 2018).

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

(Jackson et al., 2020).

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,

5
protein production. Replacing rare codons with synonymous codons frequently used by the

host cell also enhances protein production (Pardi et al., 2018).

Figure 1. mRNA components important for expressing the antigen sequence

Source. Jackson et al., (2020).

6
MECHANISM OF MRNA VACCINE

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 (Batty et al., 2020).

Traditional vaccines stimulate an antibody response by injecting either antigens,

an attenuated (weakened) virus, an inactivated  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 (Kyriakidis, 2021).

In contrast, mRNA vaccines introduce a short-lived (Hajj and Whitehead, 2017) synthetically

created fragment of the RNA sequence of a virus into the individual being vaccinated. These

mRNA fragments are taken up by dendritic cells through phagocytosis (Hajj and Whitehead,

2017). The dendritic cells use their internal machinery (ribosomes) to read the mRNA and

produce the viral antigens that the mRNA encodes (Pardi et al., 2018). The body degrades the

mRNA fragments within a few days of introduction (Anand and Stahel, 2021). 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

(Goldman, 2020). The mRNA fragments are translated in the cytoplasm and do not affect the

body's genomic DNA, located separately in the cell nucleus (Park et al., 2020).

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 (Xu et al., 2020).  Once activated, dendritic cells migrate to lymph nodes,

where they present the antigen to T cells and B cells (Xu et al., 2020). This triggers the

7
production of antibodies specifically targeted to the antigen, ultimately resulting in immunity

(Batty et al., 2020).

Figure 2. An illustration of the mechanism of action of a messenger RNA vaccine

Source. Jackson et al., (2020).

DELIVERY OF MRNA VACCINE

8
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 (Xu et al., 2020).

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 (Xu et al., 2020).

9
Figure 3. Major delivery methods and carrier molecules for mRNA vaccines

Source. Wang et al., (2021).

10
 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

(Benteyn et al., 2015).

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 (Xu et al., 2020).

 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. 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 (Benteyn et al.,

2015).

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 (Benteyn et al., 2015)

 Naked mRNA injection

Naked mRNA injection means that the delivery of the vaccine is only done in a buffer

solution (Xu et al., 2020). The first worldwide clinical studies used intradermal injections of

naked mRNA for vaccination (Wang et al., 2021). 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 (Xu et al., 2020).

11
 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 (Wang et al., 2021).

 Lipid nanoparticle vector

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 (Cooney,

2020). 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 (Cooney, 2020). Research into using lipids

to deliver siRNA to cells became a foundation for similar research into using lipids to deliver

mRNA (Cross, 2021). However, new lipids had to be invented to encapsulate mRNA strands,

which are much longer than siRNA strands (Cross, 2021).

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 (Paunovska et al., 2020). The nanoparticles can be administered to the body and

transported via multiple routes, such as intravenously or through the lymphatic system

(Cooney, 2020).

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

12
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

(Lowe, 2021). 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, (Sealy, 2021). 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," (Weiss, 2021). and set up a computer system

to regulate flow and pressure through the mixers (Sealy, , 2021).

13
Figure 4. Assembly of RNA lipid nanoparticle

Source. Buschmann, (2021)

CATEGORIES OF MRNA VACCINE

The two main categories of mRNA vaccines use either non-amplifying (conventional) mRNA

or self-amplifying mRNA. The first two approved mRNA vaccines (Pfizer–BioNTech and

Moderna COVID-19 vaccines) use non-amplifying mRNA. Both mRNA types continue to be

investigated as vaccine methods against other potential pathogens and cancer cells (Blakney

et al., 2020).

14
Figure 5. Mechanism of non-amplifying and self-amplifiying mRNA vaccines

Source. Tregoning, (2021).

 Non-amplifying mRNA

Conventional mRNA vaccines use a non-amplifying mRNA construct (Wang et al.,

2021) Non-amplifying mRNA has only one open reading frame which codes for the antigen

protein of interest (Deering et al., 2016). The total amount of mRNA used by the cell is equal

to the amount of mRNA delivered by the vaccine. In a conventional mRNA vaccine, dosage

strength is limited by the amount of mRNA that can be delivered by the vaccine (Deering et

al., 2016).

 Self-amplifying mRNA

Self-amplifying mRNA (saRNA) vaccines are similar to conventional mRNA vaccines, with

the exception, that saRNA vaccines also self-replicate their mRNA (Versteeg, 2019).  The

self-amplifying mRNA has two open reading frames. The first open reading frame, like

conventional mRNA, codes for the antigen protein of interest. The second open reading

frame codes for an RNA-dependent RNA polymerase which self-replicates the mRNA

construct in the cell and creates multiple self-copies. This allows smaller quantities of mRNA

to be used for vaccine delivery (Versteeg, 2019). The mechanisms and consequently the

15
evaluation of self-amplifying mRNA may be different, as self-amplifying mRNA is

fundamentally different by being a much bigger molecule in size (Verbeke, 2019).

Different saRNA vaccines are being researched, including development of a malaria vaccine

(Bloom et al., 2021). Gritstone bio started in 2021 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 (Knapton and Sarah, 2021).

ADVANTAGES OF MRNA VACCINE

 Traditional Vaccine

mRNA vaccines offer specific advantages over traditional vaccines (Pardi et al.,

2018). Because mRNA vaccines are not constructed from an active 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 (PHG Foundation, 2019). Another biological advantage of mRNA

vaccines is that since the antigens are produced inside the cell, they stimulate cellular

immunity, as well as humoral immunity (Dolgin, 2020).

mRNA vaccines have the production advantage that they can be designed swiftly. Moderna

designed their mRNA-1273 vaccine for COVID-19 in 2 days (Dolgin, 2020). 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 (Sealy, 2021).

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

16
than traditional flu and polio vaccines (Dolgin, 2021). 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. The majority of the days needed for each production run are allocated to rigorous

quality control at each stage (Sealy, 2021).

 DNA vaccines

In addition to sharing the advantages of theoretical DNA vaccines over established

traditional vaccines, mRNA 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 (Pardi et al., 2018).

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

(Rabson, 2021). 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 (Hajj and Whitehead, 2021). 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 (Pardi et al., 2018).

DISADVANTAGES OF MRNA VACCINE

 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–

17
BioNTech's BNT162b2 mRNA vaccine has to be kept between −80 and −60 °C (−112 and

−76 °F) (Vogel et al., 2020). Moderna says their mRNA-1273 vaccine can be stored between

−25 and −15 °C (−13 and 5 °F), (Simmons, 2020 which is comparable to a home freezer, and

that it remains stable between 2 and 8 °C (36 and 46 °F) for up to 30 days (Simmons,

2020). 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. Several platforms are being studied that may allow storage at higher

temperatures (Pardi et al., 2018).

 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 mRNA vaccines

(Pardi et al., 2018). 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 (Kramp and Elders, 2017).

Strong but transient reactogenic effects were reported in trials of novel COVID-19 mRNA

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 (Kuchler, 2020)

18
19
Figure 6. Advantages and disadvantages of different types of vaccine platforms

Source. Tregoning, (2020).

EFFICACY OF MRNA VACCINE

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 (Wadman, 2020).

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

(Wadman, 2020).

HESITANCY TO MRNA VACCINE

There is misinformation implying that mRNA vaccines could alter DNA in the nucleus

(Kwon, 2020) mRNA in the cytosol is very rapidly degraded before it would have time to

20
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 (Carmichael

and Goodman, 2020).

PFIZER–BIONTECH COVID-19 VACCINE

The Pfizer–BioNTech COVID-19 vaccine (INN: tozinameran), sold under the brand

name Comirnaty, (FDA, 2021) 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

(Browne, 2020). It is authorized for use in people aged five years and older in some

judrisdictions (CDC, 2021) twelve years and older in some jurisdictions, and for people

sixteen years and older in other jurisdictions, (EMA, 2021) to provide protection

against COVID-19, caused by infection with the SARS-CoV-2 virus (FDA, 2021). 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 (Walsh et al., 2020). Initial advice indicated that

vaccination required two doses given 21 days apart, (Walsh et al., 2020) but the interval was

later extended to up to 42 days in the US, (CDC, 2021). and up to four months in Canada

(Public Health Agency Canada, 2021).

Most side effects are mild to moderate in severity and are gone within a few days (CDC,

2021) The most common include mild to moderate pain at the injection site, fatigue,

21
and headaches (CDC, 2021). Reports of serious side effects, such as allergic reactions, are

very rare and no long-term complications have been reported (CDC, 2021).

On 23 August 2021, the Pfizer–BioNTech vaccine became the first COVID-19 vaccine to

be approved in the United States by the Food and Drug Administration (FDA) for those aged

sixteen years and older (FDA, 2021).

22
Plate 1. A vial of the Pfizer–BioNTech COVID-19 vaccine for the U.S. market

Source. Jackson (2020).

MANUFACTURING OF PFIZER AND BIONTECH VACCINE

23
Pfizer and BioNTech are manufacturing the vaccine in their own facilities in the United

States and in Europe. The license to distribute and manufacture the vaccine in China was

purchased by Fosun, alongside its investment in BioNTech (Thomas, 2020).

Manufacturing the vaccine requires a three-stage process. The first stage involves

the molecular cloning of DNA plasmids that code for the spike protein by infusing them

into Escherichia coli bacteria. For all markets, this stage is conducted in the United States

(Rabson, 2021) at a small Pfizer pilot plant in Chesterfield, Missouri (Gray, 2020) (near St.

Louis). After four days of growth, the bacteria are killed and broken open, and the contents of

their cells are purified over a week and a half to recover the desired DNA product. The DNA

is bottled and frozen for shipment. Safely and quickly transporting the DNA at this stage is so

important that Pfizer has used its company jet and helicopter to assist (Johnson, 2020).

The second stage is being conducted at a Pfizer plant in Andover, Massachusetts, (Hughes,

2020) in the United States, and at BioNTech's plants in Germany (Rabson, 2021). The

DNA is used as a template to build the desired mRNA strands, (Johnson, 2020) which takes

about four days (Rabson, 2021). Once the mRNA has been created and purified, it is frozen

in plastic bags about the size of a large shopping bag, of which each can hold up to 10 million

doses. The bags are placed on trucks which take them to the next plant (Johnson, 2020).

The third stage is being conducted at Pfizer plants in Portage, Michigan (Shamu,

2020) (near Kalamazoo) in the United States, and Puurs in Belgium. This stage involves

combining the mRNA with lipid nanoparticles, then filling vials, boxing vials, and freezing

them (Rabson, 2021). Croda International subsidiary Avanti Polar Lipids is providing the

requisite lipids (Shamus, 2020). As of November 2020, the major bottleneck in the

manufacturing process is combining mRNA with lipid nanoparticles (Rabson, 2021). At this

stage, it takes only four days to go from mRNA and lipids to finished vials, but each lot must

24
then spend several weeks in deep-freeze storage while undergoing verification against 40

quality-control measures (Rabson, 2021).

Before May 2021, (Panetta, 2021) the Pfizer plant in Puurs was responsible for all vials for

destinations outside the United States (Rabson, 2021) Therefore, all doses administered in the

Americas outside of the United States before that point in time required at least

two transatlantic flights (one to take DNA to Europe and one to bring back finished vaccine

vials (Rabson, 2021).

In February 2021, BioNTech announced it would increase production by more than 50% to

manufacture 2 billion doses in 2021,  raised again at the end of March to 2.5 billion doses in

2021 (CDC, 2021).

25
Plate 2. Boxes containing the COVID-19 vaccines at the Pfizer factory in Puurs

Source. Jackson (2020)

26
Plate 3. A Pfizer employee putting dry ice in a box to protect the COVID-19 vaccines during

transport at the Puurs factory

Source. Jackson (2020)

Plate 3. Inside view of Pfizer factory in Puurs


27
Source. Jackson (2020)

MEDICAL USES OF MRNA VACCINE

The third stage is being conducted at Pfizer plants in Portage, Michigan (Shamus,

2020) (near Kalamazoo) in the United States, and Puurs in Belgium. This stage involves

combining the mRNA with lipid nanoparticles, then filling vials, boxing vials, and freezing

them (Shamus, 2021) Croda International subsidiary Avanti Polar Lipids is providing the

requisite lipids (Hughes, 2020). As of November 2020, the major bottleneck in the

manufacturing process is combining mRNA with lipid nanoparticles. At this stage, it takes

only four days to go from mRNA and lipids to finished vials, but each lot must then spend

several weeks in deep-freeze storage while undergoing verification against 40 quality-control

measures (Shamus, 2020).

ADVERSE EFFECTS OF MRNA VACCINE

Most side effects of the Pfizer–BioNTech COVID-19 vaccine are mild to moderate in

severity and are gone within a few days (FDA, 2021). They are similar to other adult vaccines

and are normal signs that the body is building protection to the virus. During clinical trials,

the common side effects affecting more than 1 in 10 people are (in order of frequency): pain

28
and swelling at the injection site, tiredness, headache, muscle aches, chills, joint pain, and

fever (EMA, 2021). Fever is more common after the second dose (EMA, 2021).

 Allergy

Documented hypersensitivity to polyethylene glycol (PEG) (a very rare allergy) is listed as a

contraindication to the COVID-19 Pfizer vaccine (Rutkowski et al., 2021). Severe allergic

reaction has been observed in approximately eleven cases per million doses of vaccine

administered (EMA, 2021). According to a report by the US Centers for Disease Control and

Prevention, 71% of those allergic reactions happened within 15 minutes of vaccination and

mostly (81%) among people with a documented history of allergies or allergic reactions

(CDC, 2021). The UK's Medicines and Healthcare products Regulatory Agency (MHRA)

advised on 9 December 2020 that people who have a history of "significant" allergic reaction

should not receive the Pfizer–BioNTech COVID-19 vaccine (Bostok, 2020).

 Myocarditis

According to Israel's Ministry of Health there is a probable relationship between the second

dose and myocarditis in a small group of 16–30-year-old men (Heller, 2021). Between

December 2020 and May 2021, there were 55 cases of myocarditis per 1 million people

vaccinated, 95% of which were classified as mild (Heller, 2021). Since April 2021, increased

cases of myocarditis and pericarditis have been reported in the United States in about 13 per 1

million young people, mostly male and over the age of 16, after vaccination with the Pfizer–

BioNTech or the Moderna vaccine (CDC, 2021). Most affected individuals recover quickly

with adequate treatment and rest (CDC, 2021).

CONCLUSIONS

mRNA vaccines represent a promising alternative to conventional vaccine approaches

because of their high potency, capacity for rapid development and potential for low-cost

29
manufacture and safe administration. However, their application has until recently been

restricted by the instability and inefficient in vivo delivery of mRNA. Recent technological

advances have now largely overcome these issues, and multiple mRNA vaccine platforms

against infectious diseases like Covid-19 have demonstrated encouraging results in both

animal models and humans.

RECOMMENDATIONS

Decades of progress in mRNA design and nucleic acid delivery technology, together with the

discovery of novel antigen targets have made mRNA vaccines an extraordinary tool for

combating emerging pandemics and existing infectious diseases. The first two mRNA

vaccines, which were developed at revolutionary speed to fight SARS-CoV-2, have exceeded

expectations and offer hope that the COVID-19 pandemic will end. Furthermore, these

vaccines have elevated LNPs and RNA therapy from small-market products for niche

diseases to a prophylactic treatment deployed successfully in large swathes of the population.

The resultant abundance of positive safety and efficacy data, together with a proven path to

regulatory approval, leave us optimistic that mRNA therapeutics will transform modern

medicine’s approach to vaccination, cancer immunotherapy protein replacement therapy and

beyond.

SUMMARY

The potential advantages of mRNA as a vaccine range from the discovery of immunogens to

rapid response manufacturing. Currently, the field is pursuing two approaches: non-

replicating and self-replicating constructs. A number of quality attributes, that dictate stability

and efficiency of expression, continue to be an intense area of development. It is widely

recognized that the delivery of the mRNA into the cytoplasm is equally important to

successfully elicit a robust and durable immunity. As a result, much progress has been

30
achieved with considerable focus on novel ionizable lipid formulations and the next

generation of delivery systems. The major breakthrough of mRNA Vaccine research is Pfizer

and BioNtech Covid-19 Vaccine which has been very efficacious against the pandemic

disease called Covid-19.

REFERENCE

Anand, P., and Stahel, V.P. (2021).

 "Review the safety of Covid-19 mRNA vaccines: a review". Patient Safety in

Surgery. 15 (1): 20.

Bostock, N. (2020).

"MHRA warning after allergic reactions in NHS staff given COVID-19 vaccine"

Bloom, K., van den Berg, F., Arbuthnot, P. (2021).

 "Self-amplifying RNA vaccines for infectious diseases". Gene Therapy. 28 (3–4):

117–129. 

Batty, C.J., Heise, M.T., Bachelder, E.M., Ainslie, K.M. (2020). 

31
"Vaccine formulations in clinical development for the prevention of severe acute

respiratory syndrome coronavirus 2 infection". Advanced Drug Delivery

Reviews. 169: 168–89.

Buschmann, M.D.; Carrasco, M.J.; Alishetty, S.; Paige, M.; Alameh, M.G., and Weissman,

D.

(2020) Nanomaterial Delivery Systems for mRNA Vaccines. Vaccines (2021), (9),

65. 

Benteyn, D., Heirman, C., Bonehill, A., Thielemans, K., and Breckpot, K. (2015).

"mRNA-based dendritic cell vaccines". Expert Review of Vaccines. 14 (2): 161–76.

Boseley, S., and Halliday, J. (2020).

 "UK approves Pfizer/BioNTech Covid vaccine for rollout next week". The

Guardian. 

Browne, R. (2020).

 "What you need to know about BioNTech – the European company behind Pfizer's

Covid-19 vaccine". CNBC.

Blakney, Anna, K.; I.p., Shell; Geall., and Andrew, J. (2021).

 "An Update on Self-Amplifying mRNA Vaccine Development". Vaccines. 9 (2): 97. 

Crommelin, D.J., Anchordoquy, T.J., Volkin, D.B., Jiskoot, W., and Mastrobattista, E.

(2021). "Addressing the Cold Reality of mRNA Vaccine Stability". Journal of

Pharmaceutical Sciences. 110 (3): 997–1001.  

CDC COVID-19 Response Team (2021).

32
"Allergic Reactions Including Anaphylaxis After Receipt of the First Dose of Pfizer-

BioNTech COVID-19 Vaccine - United States, December 14-23,

2020"(PDF). MMWR. Morbidity and Mortality Weekly Report. 70 (2): 46–51. 

Carmichael, F., and Goodman, J. (2020). 

"Vaccine rumours debunked: Microchips, 'altered DNA' and more" (Reality Check).

BBC.

Cross, R. (2021).

 "Without these lipid shells, there would be no mRNA vaccines for COVID-

19". Chemical & Engineering News.

Cooney, E. (2020).

 "How nanotechnology helps mRNA Covid-19 vaccines work". Stat. 

CDC (2021).

 "Information about the Pfizer-BioNTech COVID-19 Vaccine". U.S. Centers for

Disease Control and Prevention (CDC). 

Deering, R.P., Kommareddy, S., Ulmer, J.B., Brito, L.A., and Geall, A.J. (2016). "Nucleic

acid

vaccines: prospects for non-viral delivery of mRNA vaccines". Expert Opinion Drug

Delivery. 11 (6): 885–99. 

Dolgin, E. (2020).

"COVID-19 vaccines poised for launch, but impact on pandemic unclear". Nature

Biotechnology. 

 EMA Press release. (28 May 2021).


33
"First COVID-19 vaccine approved for children aged 12 to 15 in EU". European

Medicines Agency

FDA Approves First COVID-19 Vaccine"(Press release) (2021).

U.S. Food and Drug Administration (FDA). (23 August 2021).

Goldman, B. (2020). "How do the new COVID-19 vaccines work?"

 Scope. Stanford Medicine.

Gray, B. (2020).

"Pfizer's Chesterfield workforce playing a key role in coronavirus vaccine

development". St. Louis Post-Dispatch. 

Hajj, K.A., and Whitehead, K.A. (2017).

 "Tools for translation: non-viral materials for therapeutic mRNA delivery". Nature

Reviews Materials. 2 (10): 17056.

Heller, J. (2021).

"Israel sees probable link between Pfizer vaccine and myocarditis cases". Reuters. 

Hughes, M. (2020). "Andover's piece of the vaccine: Pfizer". The Eagle-Tribune. 

Jackson, Nicholas, A. C.; Kester, Kent E.; Casimiro, Danilo; Gurunathan, Sanjay; and

DeRosa,

Frank, (2020). "The promise of mRNA vaccines: a biotech and industrial

perspective". NPJ Vaccines. 5 (1): 11.

Johnson, C.Y. (2020). 

34
"A vial, a vaccine and hopes for slowing a pandemic – how a shot comes to be". The

Washington Post. 

Knapton, and Sarah (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. 

Kramps, T., and Elders, K. (2017).

"Introduction to RNA Vaccines". RNA Vaccines: Methods and Protocols. Methods in

Molecular Biology. 1499. pp. 1–11. 

Kowalski, P.S., Rudra, A., Miao, L., Anderson, D.G. (2019).

 "Delivering the Messenger: Advances in Technologies for Therapeutic mRNA

Delivery". Molecular Therapy. 27 (4): 710–28. 

Kuchler, H. (2020).

 "Pfizer boss warns on risk of fast-tracking vaccines". Financial Times. 

Kwon, D. (2020). "The Promise of mRNA Vaccines". The Scientist.

Lowe, D. (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.

Mullin, R. (2020).

"Pfizer, Moderna ready vaccine manufacturing networks". Chemical & Engineering

News. Washington, D.C.: American Chemical Society.


35
Paunovska, K., Sago, C.D., Monaco, C.M., Hudson. W.H., Castro, M.G., and Rudoltz, T.G.

(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.

Public Health Agency of Canada (2021).

Recommendations on the use of COVID-19 vaccines (PDF). Health

Canada (Report). 

PHG Foundation (2019). "RNA vaccines: an introduction". University of Cambridge. 

Park, K.S., Sun, X, Aikins, M.E., and Moon, J.J. (2020).

 "Non-viral COVID-19 vaccine delivery systems". Advanced Drug Delivery

Reviews. 169: 137–51.

Pardi, Norbert; Hogan, Michael J.; Porter, Frederick W.; Weissman, and Drew (2018).

 "mRNA vaccines — a new era in vaccinology". Nature Reviews Drug

Discovery. 17 (4): 261–279.

Pardi, N., and Weissman, D. (2016).

"Nucleoside Modified mRNA Vaccines for Infectious Diseases". RNA Vaccines.

Methods in Molecular Biology. 1499. Springer New York. pp. 109–21. 

Rabson, M. (2021).

 "From science to syringe: COVID-19 vaccines are miracles of science and supply

chains". CTV News. Bell Media.

Rutkowski, K., Mirakian, R., Till, S., Rutkowski, R., and Wagner, A. (2021).

36
"Adverse reactions to COVID-19 vaccines: A practical approach". Clinical and

Experimental Allergy. 51 (6): 770–777. 

Sealy, A. (2021).

 "Manufacturing moonshot: How Pfizer makes its millions of Covid-19 vaccine

doses". CNN.

Shamus, K.J. (2020). 

"Historic journey: Pfizer prepares to deliver 6.4 million doses of COVID-19

vaccines". Detroit Free Press.

Thomas, K., Gelles, D., Zimmer, C. (2020).

Pfizer's early data shows vaccine is more than 90% effective". The New York Times. 

Tregoning, J.S., Brown, E.S., Cheeseman, H.M., Flight, K.E., Higham, S.L., Lemm, N.-M.,

Pierce, B.F., Stirling, D.C., Wang, Z. and Pollock, K.M. (2021), Vaccines for

COVID-19. Clinical. Experiment. Immunology., 202: 162-192. 

U.S. Food and Drug Administration (FDA) (Press release) (2021).

FDA Takes Additional Action in Fight Against COVID-19 By Issuing Emergency

Use Authorization for Second COVID-19 Vaccine".

Verbeke, R., Lentacker, I., De Smedt, S.C., and Dewitte, H. (2019).

 "Three decades of messenger RNA vaccine development". Nanotechnology

Today. 28: 100766. 

Vogel, A.B., Lambert, L., Kinnear, E., Busse, D., Erbar, S., and Reuter, K.C. (2018).

37
 "Self-Amplifying RNA Vaccines Give Equivalent Protection against Influenza to

mRNA Vaccines but at Much Lower Doses". Molecular Therapy. 26 (2): 446–55. 

Wadman, M. (2020).

 "Public needs to prep for vaccine side effects". Science. 370 (6520): 1022.

Wang, Y., Zhang, Z., Luo, J., Han, X., Wei, Y., and Wei, X. (2021).

 "mRNA vaccine: a potential therapeutic strategy". Molecular Cancer. 20 (1): 33. 

Weise, E. and Weintraub, K. (2021).

 "Race to the Vaccine: A COVID-19 vaccine life cycle: from DNA to doses". USA

Today. Gannett.

Walsh, E.E., Frenck, R.W., Falsey, A.R., Kitchin, N., Absalon, J., and Gurtman, A. (2020)

. "Safety and Immunogenicity of Two RNA-Based Covid-19 Vaccine

Candidates". The New England Journal of Medicine. 383 (25): 2439–50. 

Xu, Shuqin; Yang, Kunpeng; Li, Rose; and Zhang, Lu. (2020).

"mRNA Vaccine Era—Mechanisms, Drug Platform and Clinical

Prospection". International Journal of Molecular Sciences. 21 (18): 6582. 

38

You might also like