My First Book Chapter
My First Book Chapter
My First Book Chapter
Volume 50
Recent titles in the series
Volume 26 Yeast Gene Analysis
AJP Brown and MF Tuite
Volume 27 Bacterial Pathogenesis
P Williams, J Ketley and GPC Salmond
Volume 28 Automation
AG Craig and JD Hoheisel
Volume 29 Genetic Methods for Diverse Prokaryotes
MCM Smith and RE Sockett
Volume 30 Marine Microbiology
JH Paul
Volume 31 Molecular Cellular Microbiology
P Sansonetti and A Zychlinsky
Volume 32 Immunology of Infection, 2nd edition
SHE Kaufmann and D Kabelitz
Volume 33 Functional Microbial Genomics
B Wren and N Dorrell
Volume 34 Microbial Imaging
T Savidge and C Pothoulakis
Volume 35 Extremophiles
FA Rainey and A Oren
Volume 36 Yeast Gene Analysis, 2nd edition
I Stansfield and MJR Stark
Volume 37 Immunology of Infection
D Kabelitz and SHE Kaufmann
Volume 38 Taxonomy of Prokaryotes
Fred Rainey and Aharon Oren
Volume 39 Systems Biology of Bacteria
Colin Harwood and Anil Wipat
Volume 40 Microbial Synthetic Biology
Colin Harwood and Anil Wipat
Volume 41 New Approaches to Prokaryotic Systematics
Michael Goodfellow, Iain Sutcliffe, and Jongsik Chun
Volume 42 Current and Emerging Technologies for the Diagnosis of Microbial Infections
Andrew Sails and Yi-Wei Tang
Volume 43 Imaging Bacterial Molecules, Structures and Cells
Colin Harwood and Grant J. Jensen
Volume 44 The Human Microbiome
Colin Harwood
Volume 45 Microbiology of Atypical Environments
Volker Gurtler and Jack T. Trevors
Volume 46 Nanotechnology
Volker Gurtler, Andrew S. Ball and Sarvesh K. Soni
Volume 47 Methods in Microbiology
Charles S. Pavia and Volker Gurtler
Volume 48 Methods in Microbiology
Volker Gurtler
Volume 49 Methods in Silkworm Microbiology
Volker Gurtler and Gangavarapu Subrahmanyam
Methods in Microbiology
Volume 50
Covid-19: Biomedical
Perspectives
Edited by
Charles S. Pavia
Department of Biomedical Sciences,
NYIT College of Osteopathic Medicine,
New York Institute of Technology, Old Westbury;
Division of Infectious Diseases,
New York Medical College, Valhalla,
NY, United States
Volker Gurtler
School of Science,
College of Science Engineering and Health,
RMIT University, Bundoora, VIC, Australia
Academic Press is an imprint of Elsevier
125 London Wall, London, EC2Y 5AS, United Kingdom
The Boulevard, Langford Lane, Kidlington, Oxford OX5 1GB, United Kingdom
525 B Street, Suite 1650, San Diego, CA 92101, United States
50 Hampshire Street, 5th Floor, Cambridge, MA 02139, United States
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about
the Publisher’s permissions policies and our arrangements with organizations such as the Copyright
Clearance Center and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/
permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher
(other than as may be noted herein).
Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden
our understanding, changes in research methods, professional practices, or medical treatment may
become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and
using any information, methods, compounds, or experiments described herein. In using such information
or methods they should be mindful of their own safety and the safety of others, including parties for whom
they have a professional responsibility.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any
liability for any injury and/or damage to persons or property as a matter of products liability, negligence or
otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the
material herein.
ISBN: 978-0-323-85061-2
ISSN: 0580-9517 (Series)
vii
viii Contents
xiii
xiv Contributors
xvii
This page intentionally left blank
CHAPTER
Sensitive methods
for detection of
SARS-CoV-2 RNA
1
Xi Chen* and Simin Xia
The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology, Harbin,
Heilongjiang Province, People’s Republic of China
*Corresponding author: e-mail address: [email protected]
1 Introduction
Since December 2019, an outbreak of COVID-19 caused by severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) virus occurred and soon spread to the entire
world (Wang, Horby, Hayden, & Gao, 2020). The COVID-19 pandemic has led to a
dramatic loss of human life worldwide and presents an unprecedented challenge to
publish health and food systems; the global economic growth was largely slowed
down and social activities were greatly disrupted. As of July 1, 2021, COVID-19
has infected over 182 million people worldwide with over 3.9 million reported
casualties. COVID-19 is caused by an RNA virus named severe acute respiratory
syndrome coronavirus 2 (Chen et al., 2020). SARS-CoV-2 virus is a coronavirus
belonging to the beta family of coronaviruses that also includes SARS-CoV, Middle
East respiratory syndrome coronavirus (MERS-CoV), human coronavirus OC43
(HCoV-OC3), and human coronavirus HKU1 (HCoV-HKU1). These beta-viruses
are enveloped, positive-sense, single-stranded RNA viruses of zoonotic origin. As
for SARS-CoV-2, this virus contains four structural proteins, namely the envelope
protein (E), spike protein (S), membrane protein (M), and the nucleocapsid
(N) protein (Fig. 1). The S, M, and E proteins form the envelope of the virus while
the N protein is associated with the single-stranded RNA genome forming nucleo-
capsid inside the envelope.
According to initial studies, this disease caused by SARS-CoV-2 showed a high
transmissibility with a basic reproduction number R0 ¼ 1.4–5.5 (WHO, 2020; Zhao
et al., 2020), likely to be below 5 and above 3 (Chen, 2020) and therefore it is highly
desired to perform early testing of SARS-CoV-2 for timely screening of infected
individuals and to stop the spread of this disease from a location to its surroundings.
Because detection sensitivity is the key to reduce false negative results, a detection
method with a high level of sensitivity can minimize non-diagnosed infected
individuals and reduce the chance of further cross transmission (Fig. 2).
Methods in Microbiology, Volume 50, ISSN 0580-9517, https://doi.org/10.1016/bs.mim.2021.06.001
Copyright © 2022 Elsevier Ltd. All rights reserved.
1
2 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
FIG. 1
Schematic illustration of the structure of coronavirus SARS-CoV-2 and its single-stranded
RNA genome. The sequence information of the single-stranded RNA genome of SARS-CoV-2
serves as the basis for the development of nucleic acid-based diagnosis.
FIG. 2
Sensitive detection methods will be highly beneficial to prevent the spread of COVID-19. High
sensitivity of diagnostic methods ensures low false negative results and therefore reduces the
latent cross transmission of non-diagnosed but actually infected individuals to their closely
contacted people.
2 General approaches for the detection of SARS-CoV-2 3
hence was not subjected to intensive evaluation (Corman et al., 2020). Another
RT-PCR based approach for the molecular diagnosis of SARS-CoV-2 introduced
in the very beginning of the pandemic detects the ORF1b and N-gene of SARS-
CoV-2 with a sensitivity up to 10 copies per reaction using none-RNA testing
sample (Chu et al., 2020).
FIG. 3
The general principle of recombinase polymerase amplification. Step I: recombinase and
primer form complexes and target homologous DNA; step II: strand exchange forms a D-loop;
herein, the brownish and blue-coloured arrows refer to the forward and reverse primers,
respectively, upon annealing with their templates; step III: polymerase initiates synthesis; step
IV: parental strands separate & synthesis continues; herein, the hollow boxed arrows refer to
the directions of the polymerization reaction; step V: two duplexes form. Abbreviation(s):
SSB, single-stranded DNA binding protein.
primer in the presence of dNTPs (step IV & V). Cyclic repetition of this process
results in the exponential amplification of a DNA (Fig. 3) (Piepenburg et al., 2006).
In order to employ RPA for the amplification of RNA, additional reverse tran-
scriptase was added in order to convert RNA to cDNA for subsequent RPA ampli-
fication. In addition, since RNA is a much less stable species compared to DNA
and is highly prone to degradation by the ubiquitously existing RNase, RNase inhib-
itor protein was added into the RT-RPA reaction. In order to facilely detect the
amplification product, a probe and a nuclease was used. Moreover, creatine kinase,
4 Sensitive detection of SARS-CoV-2 via RT-RPA 7
phosphocreatine and ATP were needed to generate energy for the reaction. Therefore,
a practical RT-RPA based detection reaction requires at least seven enzymes/proteins
including: (i) strand-displacing DNA polymerase, (ii) recombinase, (iii) recombination-
mediator protein (RMP), (iv) single-strand DNA binding protein, (v) RNase inhibitor,
(vi) creatine kinase, and (vii) nuclease; in addition, multiple necessary reagents are
also required like dNTPs, creatine, the crowding agent—polyethylene glycol, the
activator Mg(OAc)2, the forward and reverse primers, and a probe. For the most re-
liable test, viral RNA samples need to be purified via a standard RNA purification
process rather than using a non-purified viral sample. The list of proteins, enzymes,
reagents necessary for conducting an RT-RPA reaction with their recommended con-
centrations are summarized in Table 1 (Chen et al., 2020). The concentrations of
primers, probes, RNase inhibitor and nucleases used in our hand for exo-probe,
nfo-probe and multiplexing RT-RPA reaction have also been given (Table 1).
Note: Other components include (i) RPA enzymes (120 ngμL1 T4 UvsX recombinase, 60 ngμL1 T4
UvsY recombination-mediator protein, 600 ngμL1 T4 gp32 single-stranded binding protein,
30 ngμL1 Bsu or 8.6–12.8 μg Sau DNA strand-displacing polymerase), (ii) energy-supply system
constituents (50 mM phosphocreatine, 100 ngμL1 creatine kinase, and 3 mM ATP), (iii) 200 μM each
dNTPs, (iv) buffering constituents (typically pH 7.9 50 mM Tris/100 mM KOAc), (v) reducing agent 2 mM
DTT, (vi) crowding reagent (5% carbowax 20M), and (vii) reverse transcriptase are generally included
in the commercially available RT-RPA or RT-ERA kit with fixed concentration without the need for further
adjustment (Li, Macdonald, & von Stetten, 2020).
8 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
For the detection of viral RNA using RT-RPA, multiple detecting formats are
available. These formats include exo probe, nfo probe, fpg probe, digital (Shen
et al., 2011), nesting, microfluidics (Lutz et al., 2010), solid phase, template gener-
ation, electrochemistry, colorimetric or SNP detection, and so on (Daher, Stewart,
Boissinot, & Bergeron, 2016; Li et al., 2020). Among all these, it seems that using
exo probe for fluorescence detection (Behrmann et al., 2020; Tu et al., 2020) and
using nfo probe coupled with lateral flow strip detection (Zheng et al., 2021) are
the most popular and hence we will mainly focus on these two formats for the
detection of SARS-CoV-2 viral RNA by RT-RPA. Herein, either approach has its
own advantages. For the detection of RNA using exo probe, first forward and reverse
primers that are around 30–35 nucleotides long are designed; the melting tempera-
ture of an oligonucleotide is normally not a critical factor for the performance as a
primer. The primer pairs allow the amplification of a relatively short length DNA
amplicon ideally around 100–200 bp long that is most suited for RPA amplification.
In addition to the primer pair, an exo probe was designed for the fluorogenic
detection of the amplicon in the presence of exonuclease III (exo enzyme). The exo
probe is a modified oligonucleotide featuring an abasic nucleotide analogue—
tetrahydrofuran residue (THF, sometimes referred to as “dSpacer”) that is prone to
cleavage by exo enzyme when the nucleotide is in a double-stranded state. In addition,
the exo probe is usually around 46–52 nucleotides long, with at least 30 nucleotides
located 50 - to the THF site, and a further at least 15-bp long nucleotide located
30 - to the THF site. A blocking group, such as a phosphate, a C3-spacer, a biotin-
TEG or an amine, is situated at its 30 side so that the exo probe cannot act as a primer
and block the probe from polymerase extension. The entire exo probe is able to
specifically hybridize with one chain of the amplicon (THF needs to be counted
as one nucleotide residue). In order to allow this probe to detect the amplicon in
a fluorogenic way, exo probe features a flanking dT-fluorophore (e.g. fluorescein)
on one side of the THF residue and a flanking corresponding dT-quencher group
(typically a suitable Black Hole Quencher (BHQ)) on the other side of the THF site.
In such a way, the dT-fluorophore is temporally quenched by its FRET quencher
(e.g. BHQ1) and the probe will be in a none or weakly fluorescent state. Once RPA
reaction produces an amplicon that can hybridize with the exo probe, the exo probe
is converted from single-stranded state to double-stranded state that allows the
THF residue to be readily cleaved by the exo III enzyme. Hence the quencher
moiety is separated from the probe and the fluorescence of the dT-fluorophore
is restored (Fig. 4). Exo probe allows sensitive detection in RT-RPA based on
the enhancement of fluorescence.
In order to make the sensitive detection approach more field-deployable with
minimal instrumentation, nfo probe could be designed in combination with the
use of lateral flow strips for direct visual detection. In this regard, nfo probe can
hybridize with the amplicon to give a bifunctional amplicon, typically with a biotin
antigenic tag at one end and a FAM antigenic tag at the other end. Similar to the exo
probe, a nfo probe is around 46–52 nucleotides long, features an abasic THF residue
in between, at least 30 nucleotides 50 to the THF site, at least a further 15 nucleotides
4 Sensitive detection of SARS-CoV-2 via RT-RPA 9
FIG. 4
Schematic view of the working principle of a typical exo probe for RT-RPA detection of viral
RNA.
located 30 to the THF side, and a blocking group (e.g. a phosphate) at the 30 side to
block the probe from polymerization. In contrast to exo probe, the nfo probe does not
necessitate any modified dT-fluorophore nor dT-quencher, but requires an affinity
biotin or FAM antigenic tag at the 50 side of the probe. In addition, the primer with
reverse direction to the nfo probe should also be modified with a different affinity tag
(e.g. a FAM tag or a biotin tag) complementary to the nfo probe at the 50 end. Once
the nfo probe hybridizes with the amplicon generated in the RPA amplification
reaction, the nfo probe turns to a double-stranded state, rendering its internal THF
residue susceptible to cleavage by the endonuclease IV, i.e. the nfo enzyme. Then
the 15 bp oligonucleotide 50 to the THF side is released, converting the probe to
a primer and initializing polymerization in the RT-RPA reaction. In the end, a bifunc-
tional amplicon featuring a FAM moiety at one end and a biotin moiety at the other
end is generated, which allows easy visual detection using lateral flow strips (Fig. 5).
Since RT-RPA reaction is highly viscous, a further dilution of the RT-RPA reaction
solution is necessary prior to lateral flow strip analysis.
In the lateral flow (LF) test using a lateral flow strip, the bifunctional amplicon
acts as a “bridge” that connects the gold nanoparticle and the antibody immobilized
in the test line so that the test line turns into a red colour, the colour of the gold nano-
particle (Fig. 6). In brief, the lateral flow strip contains two lines. The first line
which is closer to the absorption pad side is immobilized with α-biotin antibody.
The second control line is fixed with an antibody against α-FAM antibody (i.e.
α-[α-FAM-gold] antibody). A colloidal gold particle region (the gold particle is
coated with α-FAM antibody) is located below the two lines and above the sample
pad. When the sample pad of the test strip is inserted into a diluted RT-RPA reaction
solution, the colloidal gold particle will flow along with the eluent and sequentially
FIG. 5
Schematic view of the working mechanism of a nfo probe in RT-RPA for detection of
viral RNA.
FIG. 6
Schematic view of the working mechanism of lateral flow detection (LFD) for the detection of
bifunctional amplicon, i.e. amplicon with two antigenic labels, produced in RT-RPA reaction.
5 General considerations for designing an ultrasensitive RT-RPA assay 11
cross the test and the control line. Once the double-tagged amplicon is presented in
the RT-RPA reaction, it will bridge the gold particle and the α-biotin antibody in the
test line, turning the test line to a red colour. Meanwhile the control line that contains
α-[α-FAM-gold] antibody will bind the α-FAM antibody-coated gold particle,
suggesting that the LF strip is effective (Fig. 6).
to ensure that the designed primers indeed specifically detect certain species, but not
any other species. In order to screen a best Fw and Rw primer pair, different Fw
primer and Rw primers could be paired and evaluated.
FIG. 7
The WEPEAR protocol seamlessly combines reverse transcription and RPA reactions in one
tube allowing both steps to be conducted under their own optimal temperatures and prevents
the Mg2+ used in the RPA reaction from interacting with the RNA sample and inhibiting
reverse transcription.
the presence of Mg(OAc)2 catalyst) at their own optimal conditions (Fig. 7) (Xia &
Chen, 2020a, 2020b). Because the optimal temperature used for performing reverse
transcription and RPA reaction is different (although a bit close), direct combination
of reverse transcription and RPA reactions in one tube would compromise the entire
amplification, although possible. In addition, the Mg2+ ion used as a catalyst in the
PRA reaction can complex with the RNA to inhibit the transcription step. Hence it is
advantageous to physically separate the reverse transcription step and the PRA am-
plification step. On the other hand, however, it is also highly desirable to perform
reverse transcription and RPA reactions in one-tube without re-opening the lid again
after introducing the detection sample. In this case, it can simplify the testing pro-
cedure and reduce possible contamination during the amplification course, such
as aerosol contamination from the air. The WEPEAR protocol readily solved this
problem by seamlessly combining the RT step and the RPA step in one-pot without
opening the lid at all during the entire detection course (Fig. 7).
In WEPEAR protocol, the RNA sample is added into the reaction solution in a
vial containing all necessary reagents for both RT and RPA reaction aside from the
magnesium activator. In the meanwhile, a small volume of the magnesium acetate
solution catalyst is loaded onto the lid of the reaction vial; due to the surface tension
force, the small volume of the magnesium solution will still sit inside of the inner lid
after gently closing the lid. The reaction vial is first warmed to 37 °C for 60 s to finish
the reverse transcription process. Afterwards, the reaction vial is subjected to brief
centrifugation, vortex, centrifugation again to mix the magnesium catalyst into the
reaction mixture and initialization of the RPA reaction. Finally, the reaction vial is
14 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
warmed to 40 °C for 4 min as the pre-reaction step, vortexed, centrifuged again, and
warmed to 40 °C for another 26 min to complete the entire RPA reaction. Herein,
vortexing and centrifuging the reaction again after 4 min is beneficial to enhance
the sensitivity because RT-RPA reaction mixture is quite viscous and the internal
vortex step could help disperse the initially amplified amplicon in the entire reaction
mixture for a more efficient amplification (Fig. 7).
In the WEPEAR-powered RT-PRA approach, two exo probes were designed for
the detection of N-gene and S-gene. Both probes show high sensitivity for the detec-
tion of SARS-CoV-2 RNA. The exo probe for N-gene can detect four copies of RNA
in a reaction while the exo probe for S-gene is able to achieve single-copy sensitivity
in the RT-RPA reaction.
As has been mentioned above, false negative results that are usually caused by
lack of sufficient sensitivity in the detection method should be avoided. False pos-
itive results also need to be minimized for a reliable diagnostic approach. Herein, if
two genes could be simultaneously detected, many false positive results will be ex-
cluded. In our recent report, the exo probe for N-gene is green-colour emissive and
the exo probe for S-gene is red-colour emissive. Hence, it makes it possible to mul-
tiplex the RT-RPA reaction for simultaneous dual-gene detection. We achieved this
simply by reducing the concentrations of both primers and probes to around one fifth
of their original concentration. Real-time dual-gene detection using green and red
channels was readily achieved using an advanced real-time PCR machine. This result
suggested that rational adjustment/reduction of the primer and probe concentrations
is an efficient way to establish a successful multiplexing RT-RPA reaction.
In addition to the exo probes, nfo probe for both N- and S-gene were also
designed for lateral flow test without the need of a real-time PCR machine, nor a blue
light imaging plate. Hence, nfo probe combined with lateral flow strips provide a
more field-deployable way for conducting SARS-CoV-2 detection. Household
detection using this LFD method is also possible simply using a daily used thermos
cup to warm up the reaction vial. Herein, ultrasensitivity has been achieved for both
nfo probes. The sensitivity tested using in vitro transcribed RNA sample reached
four-copy per reaction.
Among other RT-RPA methods for SARS-CoV-2 detection (Table 2), enhanced
RT-RPA has been introduced which is named eRPA protocol (Qian et al., 2020). In-
ternally quenched (exo-IQ) probe was designed for the detection of SARS-CoV-2
N gene (Behrmann et al., 2020). Also, all the required detection setup and reagents
could be included in a suitcase called a “Suitcase Lab” for deployable detection of
SARS-CoV-2 (Behrmann et al., 2020). Moreover, RT-RPA can be coupled with gene
editing enzymes, e.g. Cas12a or Cas13a for viral RNA detection (Arizti-Sanz et al.,
2020; Xiong et al., 2020). RT-RPA has the following advantages over RT-PCR:
(1) the amplification proceeds faster which can be accomplished within 20–30min;
(2) RT-RPA proceeds at a constant temperature without the need of thermocyclers,
and often gives higher sensitivity. For example, there are already several reports
achieving single-copy detection sensitivity for the detection of SARS-CoV-2 using
Table 2 Summarization of recently published RT-RPA methods for sensitive detection of SARS-CoV-2 (as of February 2021).
Entry Gene Type Sequence & structure Duration Sensitivity Specificity Readout References Date
1 and N Fw TTTGGTGGACCCTCAGATTCAACTGGCAGTAAC 1 min (37 °C) 4-copy/ Specific over Green Xia and March 23,
2 Rw GAATTTAAGGTCTTCCTTGCCATGTTGAGTGAG + 30 min reaction SARS-CoV emission Chen 2020
(40 °C) with (50 μL) (2020a) and
Exo-probe TATTATTGGGTAAACCTTGGGGCCGACGTTGTT/
internal vortex Xia and
i6FAMdT/T/idSp/
Chen
A/iBHQ1dT/CGCGCCCCACTG-Phosphate
(2020b)
N Fw TTTGGTGGACCCTCAGATTCAACTGGCAGTAAC 1 min (37 °C) 4-copy/ Specific over LFD May 28,
Rw Biotin-GAATTTAAGGTCTTCCTTGCCATGT + 30 min reaction MERS-CoV 2020
TGAGTGAG (40 °C) with (50 μL) but not
internal vortex SARS-CoV
Nfo-probe 6-FAM-GCGATCAAAACAACGTCGGCCCCAAGGTT
+2 min LF test
TACC/idSp/AATAATACTGCGTCT-Phosphate
S Fw GTCTCTAGTCAGTGTGTTAATCTTACAACCAGAAC 1 min (37 °C) 1-copy/ Specific over Red Xia and May 28,
Rw CATTGGAAAAGAAAGGTAAGAACAAGTCCTGAG + 30 min reaction SARS-CoV emission Chen 2020
(40 °C) with (50 μL) (2020b)
Exo-probe CCTGCATACACTAATTCTTTCACACGTGGT
internal vortex
G/iTAMdT/T/idSp/A/iBHQ2dT/
TACCCTGACAAAGTT-Phosphate
S Fw GTCTCTAGTCAGTGTGTTAATCTTACAACCAGAAC 1 min (37 °C) 4-copy/ Specific over LFD
Rw Biotin-CATTGGAAAAGAAAGGTAAGAACAAGT + 30 min reaction SARS-CoV,
CCTGAG (40 °C) with (50 μL) MERS-CoV
internal vortex
Nfo-probe 6-FAM-CCTGCATACACTAATTCTTTCACACGTGGT
+2 min LF test
G/idSp/TTATTACCCTGACAA-Phosphate
3 N Fw CCTCTTCTCGTTCCTCATCACGTAGTCGCAAC 21 min (42 °C) 10-copy/ Specific over Green Behrmann May 8,
Rw AGTGACAGTTTGGCCTTGTTGTTGTTGGCCTT + internal reaction SARS-CoV, emission et al. (2020) 2020
vortex (57 μL) MERS-CoV,
Exo- CCTGCTAGAATGGCTGGCAATGGCGGTGA/
OC43/229E/
Probe idFAMdT/idSp/C/iBMNQ535dT/
NL63
TGCTCTTGCTTTGC-C3
4 S Fw CTTCAACCTAGGACTTTTCTATTAAAATATAATG 4 min + 20 min 10-copy/ Specific over Green Xue et al. May 22,
Rw GTTGGTTGGACTCTAAAGTTAGAAGTTTGATAG (39 °C) reaction OC43/229E/ emission (2020) 2020
(50 μL) NL63/HKU1
Exo-probe CCATTACAGATGCTGTAGACTGTGCACTTG
ACCC/iFAMdT/C/idSp/C/iBHQ1dT/
CAGAAACAAAGTGTACG-Phosphate
Orf1ab Fw TACGCCAAGCTTTGTTAAAAACAGTACAATTCTG 4 min + 20 min 1-copy/ Green
Rw GGCATTAACAATGAATAATAAGAATCTACAACAGG (39 °C) reaction emission
(50 μL)
Exo-probe TTGTTGGTGTACTGACATTAGATAATCAAG
ATC/iFAMdT/C/idSp/A/iBHQ1dT/
GGTAACTGGTATGATTTCG-Phosphate
5 N Fw CAGTTCAAGAAATTCAACTCCAGGCAGCAGTAG 7 min (39 °C) 10-copy/ Specific over Green Wu et al. July 29,
Rw CAGTTTGGCCTTGTTGTTGTTGGCCTTTAC + 20 min reaction OC43/229E/ emission (2020) 2020
(39 °C) (50 μL) NL63/HKU1,
influenza A/
Exo-probe CAGACATTTTGCTCTCAAGCTGGTTCAATC
B; unknown
/iFAMdT/idSp/iBHQ1dT/CAAGCAGCAGC
for SARS-
AAAG-C3
CoV
6 S Fw TCTTGTTTTATTGCCACTAGTCTCTAGTCAGT 25 min (42 °C) 3-copy/ Specific over LFD Qian et al. November
Rw FAM-GAATGTAAAACTGAGGATCTGAAAACTTTG + 3 min (90 °C) reaction SARS-CoV, (2020) 20, 2020
+ 3 min (RT) (50 μL) MERS-CoV,
+3 min LF test SuperScript 229E/HKU1
Nfo-probe Biotin-TGCATACACTAATTCTTTCACACGTGGT
IV & RNase
H added
Continued
Table 2 Summarization of recently published RT-RPA methods for sensitive detection of SARS-CoV-2 (as of February 2021).—
cont’d
Entry Gene Type Sequence & structure Duration Sensitivity Specificity Readout References Date
7 Orf1ab Fw CCAAGGTAAACCTTTGGAATTTGGTGCCAC 50 min 10-copy/μL Specific over Green Arizti-Sanz November
input SARS-CoV emission et al. (2020) 20, 2020
Rw ACTATCATCATCTAACCAATCTTCTTCTTG or LFD
Cas13a CUCUUCUUCAGGUUGAAGAGCAGCAGAA
crRNA
FQ/FB 6-FAM-rUrUrUrUrUrUrUrUrUrUrUrUrUrU-Biotin
reporter 6-FAM-rUrUrUrUrUrUrU-IABkFQ
8 Orf1ab Fw TTGCCTGGCACGATATTACGCACAACTAATGGT 20 min (42 °C) 1-copy/ Specific over Green Xiong et al. December
Rw CAAGCTGATGTTGCAAAGTCAGTGTACTCTAT + up to reaction MERS-CoV, emission (2020) 15, 2020
120 min (37 °C) (12.5 μL) SARS-CoV, or LFD
Cas12a UAAUUUCUACUAAGUGUAGAUgugcaguug
for Cas12a OC43/HKU1
crRNA guaacaucuguuac
detection
FQ/FB FAM-TTATT-BHQ1
reporter FAM-TTATT-Biotin
N Fw CTTCCTCAAGGAACAACATTGCCAAAAGGCT 20 min (42 °C) 1–10-copy/ Specific over Green
Rw TCTAGCAGGAGAAGTTCCCCTACTGCTGCCTGG + up to reaction SARS-CoV, emission
120 min (37 °C) (12.5 μL) MERS-CoV, or LFD
Cas12a UAAUUUCUACUAAGUGUAGAUuugaacugu
for Cas12a OC43/HKU1
crRNA ugcgacuacgugau
detection
FQ/FB FAM-TTATT-BHQ1
reporter FAM-TTATT-Biotin
9 RdRp Fw TATGCCATTAGTGCAAAGAATAGAGCTCGCAC 15 min 2-copy/ NOT specific Green Abd El January
Rw CAACCACCATAGAATTTGCTTGTTCCAATTAC reaction over SARS- emission Wahed et al. 20, 2021
(50 μL) CoV (2021)
Exo-probe TCCTCTAGTGGCGGCTATTGATTTCAATAA
/iBHQ1dT/idSp/iFAMdT/
TTGATGAAACTGTCTATTG-Phosphate
E Fw GAAGAGACAGGTACGTTAATAGTTAATAGCGTA 15 min 15-copy/ NOT specific Green
Rw AAAAAGAAGGTTTTACAAGACTCACGTTAACsA reaction over SARS- emission
(50 μL) CoV
Exo-probe ATCGAAGCGCAGTAAGGATGGCTAG/iBHQ1dT/
idSp/iFAMdT/AACTAGCAAGAATAC-Phosphate
N Fw CCTCTTCTCGTTCCTCATCACGTAGTCGCAAC 15 min 15-copy/ Specific over Green
Rw AGTGACAGTTTGGCCTTGTTGTTGTTGGCCTT reaction SARS-CoV emission
(50 μL)
Exo-probe TAGAATGGCTGGCAATGGCGGTGATGCTGC
/iBHQ1dT/idSp/iFAMdT/TGCTTTGCTGCTGCTT-
Phosphate
10 N Fw CTAACAAAGACGGCATCATATGGGTT 30 min (39 °C) 10-copy/ Specific over LFD Zheng et al. February
Rw FITC-GGCCTTTACCAGACATTTTGCTCTCA + LFD reaction SARS-CoV, (2021) 1, 2021
(50 μL) OC43/
Nfo-probe Biotin-CTCTTCTCGTTCCTCATCACGTAGTCGCAA
HKU1/
C/dSp/GTTCAAGAAATTCA-Phosphate
NL63/229E,
HBV human
influenza A/
B, RSV A/B,
Abbreviations: FITC, fluorescein isothiocyanate; FAM, fluorescein; 6-FAM, 6-carboxyfluorescein; C3, C3-blocking group; idFAMdT, internal FAM modified dT nucleotide; id6FAMdT, internal 6-FAM modified dT nucleotide; idTAMdT,
internal TAMRA modified dT nucleotide; idSp, internal tetrahydrofuran (THF) spacer; idBHQ1, internal BHQ1 quencher modified dT nucleotide; idBHQ2, internal BHQ2 quencher modified dT nucleotide; iBMNQ535dT, internal
BMNQ535 quencher modified dT nucleotide; s, phosphorothioate backbone.
9 Key resources table 17
RT-RPA (Xia & Chen, 2020a, 2020b; Xue et al., 2020). A comparison of the recently
reported RT-RPA detection against SARS-CoV-2 RNA is summarized in Table 2.
7 Methods section
In the following, we will detail the methods using the WEPEAR protocol for ultra-
sensitive field-deployable detection of SARS-CoV-2 RNA.
Methods section (All Using Basic RT-ERA Kit).
Continued
18 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
—cont’d
Reagent or resource Source Identifier
Basic RT-ERA reaction kit GenDx Biotech Co., Cat# KS102
Ltd.
FastPure gel DNA extraction mini kit Vazyme Biotech Cat# DC301
Co., Ltd.
Express RNA purification kit GenDx Biotech Co., Cat# NR202-50T
Ltd
Oligonucleotides
N-gene Exo Fw: LoGenBio N.A.
tttggtggaccctcagattcaactggcagt
aac
N-gene Exo Rw: LoGenBio N.A.
gaatttaaggtcttccttgccatgttgagt
gag
N-gene Exo probe: LoGenBio N.A.
tattattgggtaaaccttggggccgacgtt
gtt/i6FAMdT/t/idSp/a/iBHQ1dT/
cgcgccccactg-Phosphate
N-gene Nfo Fw: LoGenBio N.A.
tttggtggaccctcagattcaactggcagt
aac
N-gene Nfo Rw: LoGenBio N.A.
tttggtggaccctcagattcaactggcagt
aac
N-gene Nfo probe: 6- LoGenBio N.A.
FAM-gcgatcaaaacaacgtcggccccaaggttt
acc/idSp/aataatactgcgtct-Phosphate
S-gene Exo Fw: LoGenBio N.A.
gtctctagtcagtgtgttaatcttacaacc
agaac
S-gene Exo Rw: LoGenBio N.A.
cattggaaaagaaaggtaagaacaagtcct
gag
S-gene Exo probe: LoGenBio N.A.
cctgcatacactaattctttcacacgtggt
g/iTAMdT/t/idSp/A/iBHQ2dT/
taccctgacaaagtt-Phosphate
S-gene Nfo Fw: LoGenBio N.A.
gtctctagtcagtgtgttaatcttacaacc
agaac
S-gene Nfo Rw: Biotin- LoGenBio N.A.
cattggaaaagaaaggtaagaacaagtcct
gag
S-gene Nfo probe: LoGenBio N.A.
6-FAM-cctgcatacactaattctttcacacgtggt
g/idSp/ttattaccctgacaa-Phosphate
Recombinant DNA
SARS-CoV-2 N-gene plasmid LoGenBio N.A.
11 Step-by-step method details 19
—cont’d
Reagent or resource Source Identifier
SARS-CoV-2 S-gene plasmid LoGenBio N.A.
SARS-CoV N-gene plasmid LoGenBio N.A.
SARS-CoV S-gene plasmid LoGenBio N.A.
MERS-CoV N-gene plasmid LoGenBio N.A.
MERS-CoV S-gene plasmid LoGenBio N.A.
Software and algorithms
SnapGene viewer SnapGene www.snapgene.com/
snapgene-viewer
BLAST NCBI https://blast.ncbi.nlm.
nih.gov/Blast.cgi
Image J National Institutes of https://imagej.net/
Health (NIH) Downloads
Other
HybriDetect LF strips Amplification Future Cat# WLF8201
DL5000 DNA marker Vazyme Cat# MD102
100 bp DNA ladder Vazyme Cat# MD104
Agarose Biosharp Cat# BS081-100g
50 TAE buffer Alphabio Cat# A1558
SYBR safe DNA gel stain APExBio Cat# A8743
DNA sample loading buffer Alphabio Cat# A1550
Important notes:
(i) The area around PCR machine where RT-ERA amplification is conducted can
produce invisible aerosols in the air that contain the target amplicon. Hence
the aerosols could potentially contaminate the surrounding areas. As a result,
the area for setting up the RT-ERA reaction and the area for conducting the
RT-ERA amplification should be strictly separated in order to prevent the
contamination of the RT-ERA reaction solution by aerosols in the air.
(ii) Since RNase ubiquitously exists which is detrimental for RNA samples, the
whole working area should be carefully cleaned using DEPEC water and the
equipment used (e.g. mini-centrifuge and pipettes) should also be swiped using
DEPEC water prior to use; in addition, all consumables, e.g. tips, should be
autoclaved after spraying with DEPEC water prior to use.
(iii) Always wear appropriate personal protective equipment (PPE) including at
least a lab coat, goggles, gloves, and a mask; wearing of a mask is required in
order to prevent any foams/droplets that contain RNase and other contaminant to
be expelled from the mouth and nose contaminating the RT-ERA reaction.
(iv) Be highly cautious to avoid any part of your skin from touching the working
bench, equipment and consumables when performing the SARS-CoV-2
detection because skin surfaces have a lot of secretions (like oil) that contain
RNase and other contaminates.
(v) Due to the ultrahigh sensitivity of this RT-ERA test, both positive control and
negative control samples must be included in every test in order to exclude
possible false positive and false negative results.
11 Step-by-step method details 21
Important notes:
(i) The area around the PCR machine where RT-ERA amplification is conducted
can produce invisible aerosols in the air that contain the target amplicon. Hence
the aerosols could potentially contaminate the surrounding areas. As a result,
the area for setting up the RT-ERA reaction and the area for conducting the
RT-ERA amplification should be strictly separated in order to prevent the
contamination of the RT-ERA reaction solution by aerosols in the air.
(ii) Since RNase ubiquitously exists, which is detrimental for RNA samples, the
whole working area should be carefully cleaned using DEPEC water and the
equipment used (e.g. mini-centrifuge and pipettes) should also be swiped using
DEPEC water prior to use; in addition all consumables, e.g. tips, should
be autoclaved after spraying with DEPEC water prior to use.
(iii) Always wear appropriate PPE including at least a lab coat, goggles, gloves, and
mask; wearing of a mask is required in order to prevent any foams/droplets that
22 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
contain RNase and other contaminants to be expelled from the mouth and nose
contaminating the RT-ERA reaction.
(iv) Be highly cautious to avoid any part of your skin from touching the working
bench, equipment and consumables when performing the SARS-CoV-2
detection because skin surfaces have a lot of secretions (like oil) that contain
RNase and other contaminates.
(v) Due to the ultrahigh sensitivity of this RT-ERA test, both a positive control and
a negative control samples must be included in every test in order to exclude
possible false positive and false negative results.
(vi) After the LF strip is dipped into the diluted RT-ERA reaction, the results should
be read within the initial few minutes to get a reliable readout because the
test line may still gradually, although slowly, develop some light red colour
after an extended duration even for a blank or negative control sample.
12 Summary
In this chapter, we have described nucleic acid-based approaches for sensitive detec-
tion of SARS-CoV-2 with a particular focus on the recombinase polymerase ampli-
fication that belongs to the isothermal amplification category. Sensitivity is the key
to avoid false negative results and would be highly beneficial to prevent the spread of
a pandemic. Some recent reports have already pointed out the necessity to avoid in-
sensitive methods, e.g. some serological tests, for practical SARS-CoV-2 diagnosis.
According to recently published papers, more than one RT-RPA-based reports have
achieved single-copy sensitivity, the highest sensitivity in diagnosis. This is gener-
ally higher than the widely used RT-PCR methods and is clearly higher than other
testing approaches. In order to achieve a high sensitivity, primers and probe should
be rationally designed in order to achieve the best performance. Specificity should
also be taken into account when designing primers and probes for sensitive viral
RNA detection. The region within an RNA genome for RT-RPA amplification also
needs to be carefully taken into account. Unlike the direct detection of DNA using
RPA, the detection of RNA using RPA requires an additional reverse transcription
step. However, the reaction condition of reverse transcription is typically not fully
consistent with the conditions used in RPA; and moreover, some reagents used in
RPA (e.g. the catalyst Mg2+) can potentially inhibit reverse transcription. In order
to make the two steps fully compatible, whole-course encapsulated procedure for
exponential amplification from RNA (WEPEAR) protocol has been introduced
for “sample-in, results-out” one tube detection of viral RNA sample (Xia & Chen,
2020a, 2020b).
RT-RPA has several distinct advantages; however, it also has some limitations.
RT-RPA is performed under a constant temperature, in the range between room tem-
perature to 42 °C without the need for thermocyclers. As a result, highly costly real-
time PCR machine is not compulsory for performing the detection. RT-RPA could be
coupled with either fluorogenic detection using a blue light imaging plate, or lateral
flow strips without any particular imaging devices. All these features make RT-RPA
highly field-deployable and would be rather suited for grassroots clinics. In fact, we
have shown that RT-PRA coupled with LF test using thermos cups as the heating
device can be conducted in a house-hold fashion (Xia & Chen, 2020a, 2020b). An-
other key advantage of RT-RPA is the fast detection rate with a typical sample-to-
readout time in the range of 20–30 min, and even within 20 min. This is clearly
shorter than the RT-LAMP method (usually >60 min) and RT-PCR methods (typi-
cally >2 h). On the other hand, the RT-RPA reaction contains multiple components,
including several enzymes, proteins, energy-supply system, crowding agents, aside
from primers and probes. This makes a standard RT-RPA reaction relatively more
expensive. However, given that no sophisticated instrumentation is required and
that diagnosis time is much shorter, the general cost for performing an RPA assay
could still be reduced to a comparative level to other nucleic acid-based detection
approaches as suggested by some previous evaluations (Londono, Harmon, &
Polston, 2016). Along with the further development of RT-RPA, such as the digital
24 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
version, microfluidic version and so on, and the fact that there are several unique
advantages of RT-RPA (e.g. ultrahigh sensitivity), we predict RT-RPA approach will
show great potential for sensitive and field-deployable viral RNA detection in the
future.
Acknowledgement
We thank the research fund from Harbin Institute of Technology, and National Natural Science
Foundation of China (grant No. 32071410 to X.C.) for the support of this work.
References
Abd El Wahed, A., Patel, P., Maier, M., Pietsch, C., Ruster, D., Bohlken-Fascher, S., et al.
(2021). Suitcase lab for rapid detection of SARS-CoV-2 based on recombinase polymerase
amplification assay. Analytical Chemistry, 93(4), 2627–2634.
Ai, T., Yang, Z., Hou, H., Zhan, C., Chen, C., Lv, W., et al. (2020). Correlation of chest CT and
RT-PCR testing for coronavirus disease 2019 (COVID-19) in China: A report of 1014
cases. Radiology, 296(2), E32–E40.
Amanat, F., Stadlbauer, D., Strohmeier, S., Nguyen, T. H. O., Chromikova, V., McMahon, M.,
et al. (2020). A serological assay to detect SARS-CoV-2 seroconversion in humans.
Nature Medicine, 26(7), 1033–1036.
Arizti-Sanz, J., Freije, C. A., Stanton, A. C., Petros, B. A., Boehm, C. K., Siddiqui, S., et al.
(2020). Streamlined inactivation, amplification, and Cas13-based detection of SARS-
CoV-2. Nature Communications, 11(1), 5921.
Behrmann, O., Bachmann, I., Spiegel, M., Schramm, M., Abd El Wahed, A., Dobler, G., et al.
(2020). Rapid detection of SARS-CoV-2 by low volume real-time single tube reverse tran-
scription recombinase polymerase amplification using an exo probe with an internally
linked quencher (Exo-IQ). Clinical Chemistry, 66(8), 1047–1054.
Chen, J. L. (2020). Pathogenicity and transmissibility of 2019-nCoV-A quick overview and
comparison with other emerging viruses. Microbes and Infection, 22(2), 69–71.
Chen, L. J., Liu, W. Y., Zhang, Q., Xu, K., Ye, G. M., Wu, W. C., et al. (2020). RNA based
mNGS approach identifies a novel human coronavirus from two individual pneumonia
cases in 2019 Wuhan outbreak. Emerging Microbes & Infections, 9(1), 313–319.
Chen, J. S., Ma, E., Harrington, L. B., Da Costa, M., Tian, X., Palefsky, J. M., et al. (2018).
CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity.
Science, 360(6387), 436–439.
Chu, D. K. W., Pan, Y., Cheng, S. M. S., Hui, K. P. Y., Krishnan, P., Liu, Y. Z., et al. (2020).
Molecular diagnosis of a novel coronavirus (2019-nCoV) causing an outbreak of pneumo-
nia. Clinical Chemistry, 66(4), 549–555.
Compton, J. (1991). Nucleic-acid sequence-based amplification. Nature, 350(6313), 91–92.
Corman, V. M., Landt, O., Kaiser, M., Molenkamp, R., Meijer, A., Chu, D. K. W., et al. (2020).
Detection of 2019 novel coronavirus (2019-nCoV) by real-time RT-PCR. Eurosurveil-
lance, 25(3), 23–30.
Daher, R. K., Stewart, G., Boissinot, M., & Bergeron, M. G. (2016). Recombinase polymerase
amplification for diagnostic applications. Clinical Chemistry, 62(7), 947–958.
References 25
Dinnes, J., Deeks, J. J., Adriano, A., Berhane, S., Davenport, C., Dittrich, S., et al. (2020).
Rapid, point-of-care antigen and molecular-based tests for diagnosis of SARS-CoV-2 in-
fection. Cochrane Database of Systematic Reviews, 8, 1–129. https://doi.org/
10.1002/14651858.CD14013705.
Dohla, M., Boesecke, C., Schulte, B., Diegmann, C., Sib, E., Richter, E., et al. (2020). Rapid
point-of-care testing for SARS-CoV-2 in a community screening setting shows low sen-
sitivity. Public Health, 182, 170–172.
Gootenberg, J. S., Abudayyeh, O. O., Lee, J. W., Essletzbichler, P., Dy, A. J., Joung, J., et al.
(2017). Nucleic acid detection with CRISPR-Cas13a/C2c2. Science, 356(6336), 438–442.
James, A. S., & Alawneh, J. I. (2020). COVID-19 infection diagnosis: Potential impact of iso-
thermal amplification technology to reduce community transmission of SARS-CoV-2. Di-
agnostics, 10(6), 399.
Kashir, J., & Yaqinuddin, A. (2020). Loop mediated isothermal amplification (LAMP) assays
as a rapid diagnostic for COVID-19. Medical Hypotheses, 141, 109786.
Kilic, T., Weissleder, R., & Lee, H. (2020). Molecular and immunological diagnostic tests of
COVID-19: Current status and challenges. iScience, 23(8), 101406.
Li, J., Macdonald, J., & von Stetten, F. (2020). Review: A comprehensive summary of a de-
cade development of the recombinase polymerase amplification (vol 144, pg 31, 2019).
Analyst, 145(5), 1950–1960.
Londono, M. A., Harmon, C. L., & Polston, J. E. (2016). Evaluation of recombinase polymer-
ase amplification for detection of begomoviruses by plant diagnostic clinics. Virology
Journal, 13, 48.
Lutz, S., Weber, P., Focke, M., Faltin, B., Hoffmann, J., Muller, C., et al. (2010). Microfluidic
lab-on-a-foil for nucleic acid analysis based on isothermal recombinase polymerase am-
plification (RPA). Lab on a Chip, 10(7), 887–893.
Notomi, T., Okayama, H., Masubuchi, H., Yonekawa, T., Watanabe, K., Amino, N., et al.
(2000). Loop-mediated isothermal amplification of DNA. Nucleic Acids Research,
28(12), E63.
Piepenburg, O., Williams, C. H., Stemple, D. L., & Armes, N. A. (2006). DNA detection using
recombination proteins. PLoS Biology, 4(7), 1115–1121.
Qian, J., Boswell, S. A., Chidley, C., Lu, Z. X., Pettit, M. E., Gaudio, B. L., et al. (2020). An
enhanced isothermal amplification assay for viral detection. Nature Communications,
11(1), 5920.
Qin, Z., Peng, R., Baravik, I. K., & Liu, X. Y. (2020). Perspective fighting COVID-19: Inte-
grated micro- and nanosystems for viral infection diagnostics. Matter, 3(3), 628–651.
Shen, F., Davydova, E. K., Du, W. B., Kreutz, J. E., Piepenburg, O., & Ismagilov, R. F. (2011).
Digital isothermal quantification of nucleic acids via simultaneous chemical initiation of
recombinase polymerase amplification reactions on SlipChip. Analytical Chemistry,
83(9), 3533–3540.
Shen, M. Z., Zhou, Y., Ye, J. W., Al-Maskri, A. A. A., Kang, Y., Zeng, S., et al. (2020). Recent
advances and perspectives of nucleic acid detection for coronavirus. Journal of Pharma-
ceutical Analysis, 10(2), 97–101.
Tang, M. S., Hock, K. G., Logsdon, N. M., Hayes, J. E., Gronowski, A. M., Anderson, N. W.,
et al. (2020). Clinical performance of two SARS-CoV-2 serologic assays. Clinical Chem-
istry, 66(8), 1055–1062.
Tu, F., Yang, X. T., Xu, S. K., Chen, D. J., Zhou, L., Ge, X. N., et al. (2020). Development of a
fluorescent probe-based real-time reverse transcription recombinase-aided amplification
assay for the rapid detection of classical swine fever virus. Transboundary and Emerging
Diseases. https://doi.org/10.1111/tbed.13849.
26 CHAPTER 1 Sensitive methods for detection of SARS-CoV-2 RNA
Udugama, B., Kadhiresan, P., Kozlowski, H. N., Malekjahani, A., Osborne, M., Li, V. Y. C.,
et al. (2020). Diagnosing COVID-19: The disease and tools for detection. ACS Nano,
14(4), 3822–3835.
Vincent, M., Xu, Y., & Kong, H. M. (2004). Helicase-dependent isothermal DNA amplifica-
tion. EMBO Reports, 5(8), 795–800.
Walker, G. T., Fraiser, M. S., Schram, J. L., Little, M. C., Nadeau, J. G., & Malinowski, D. P.
(1992). Strand displacement amplification—An isothermal, in vitro DNA amplification
technique. Nucleic Acids Research, 20(7), 1691–1696.
Wang, J., Cai, K., He, X., Shen, X., Wang, J., Liu, J., et al. (2020). Multiple-centre clinical
evaluation of an ultrafast single-tube assay for SARS-CoV-2 RNA. Clinical Microbiology
and Infection, 26(8), 1076–1081.
Wang, C., Horby, P. W., Hayden, F. G., & Gao, G. F. (2020). A novel coronavirus outbreak of
global health concern (vol 395, pg 470, 2020). Lancet, 395(10223), 496.
Wang, L., Qian, C., Wu, H., Qian, W. J., Wang, R., & Wu, J. (2018). Technical aspects of
nicking enzyme assisted amplification. Analyst, 143(6), 1444–1453.
WHO. (2020). Statement on the first meeting of the international health regulations
(2005) Emergency committee regarding the outbreak of novel coronavirus (2019-nCoV).
Online Post.
Wong, Y. P., Othman, S., Lau, Y. L., Radu, S., & Chee, H. Y. (2018). Loop-mediated isother-
mal amplification (LAMP): A versatile technique for detection of micro-organisms. Jour-
nal of Applied Microbiology, 124(3), 626–643.
Wu, T., Ge, Y. Y., Zhao, K. C., Zhu, X. J., Chen, Y., Wu, B., et al. (2020). A reverse-
transcription recombinase-aided amplification assay for the rapid detection of N gene
of severe acute respiratory syndrome coronavirus 2(SARS-CoV-2). Virology, 549, 1–4.
Xia, S., & Chen, X. (2020a). Ultrasensitive and whole-course encapsulated field detection of
2019-nCoV gene applying exponential amplification from RNA combined with chemical
probes. ChemRixv. https://doi.org/10.26434/chemrxiv.12012789.
Xia, S., & Chen, X. (2020b). Single-copy sensitive, field-deployable, and simultaneous dual-
gene detection of SARS-CoV-2 RNA via modified RT-RPA. Cell Discovery, 6(1), 37.
Xiong, D., Dai, W. J., Gong, J. J., Li, G. D., Liu, N. S., Wu, W., et al. (2020). Rapid detection of
SARS-CoV-2 with CRISPR-Cas12a. PLoS Biology, 18(12), e3000978.
Xue, G. H., Li, S. L., Zhang, W. W., Du, B., Cui, J. H., Yan, C., et al. (2020). Reverse-
transcription recombinase-aided amplification assay for rapid detection of the 2019 novel
coronavirus (SARS-CoV-2). Analytical Chemistry, 92(14), 9699–9705.
Zhao, Y. X., Chen, F., Li, Q., Wang, L. H., & Fan, C. H. (2015). Isothermal amplification of
nucleic acids. Chemical Reviews, 115(22), 12491–12545.
Zhao, S., Lin, Q. Y., Ran, J. J., Musa, S. S., Yang, G. P., Wang, W. M., et al. (2020). Prelim-
inary estimation of the basic reproduction number of novel coronavirus (2019-nCoV) in
China, from 2019 to 2020: A data-driven analysis in the early phase of the outbreak.
International Journal of Infectious Diseases, 92, 214–217.
Zheng, Y. Z., Chen, J. T., Li, J., Wu, X. J., Wen, J. Z., Liu, X. Z., et al. (2021). Reverse tran-
scription recombinase-aided amplification assay with lateral flow dipstick assay for rapid
detection of 2019 novel coronavirus. Frontiers in Cellular and Infection Microbiology, 11,
613304.
CHAPTER
1 Introduction
Many diseases have a seasonal cycle occurring once or multiple times a year, or once
every several years. This regular temporal behaviour is observed because the emer-
gence of infectious diseases depends on a multiplicity of factors, including the
seasons, weather and geography, the number of susceptible individuals in a popula-
tion, the severity of the pathogen, host physiology and phenology, and the genomic
makeup of the virus, which can convert a non-infectious pathogen to one that is
infectious for a specific or general population (Dowell, 2001; Martinez, 2018).
The transmission of viral infections associated with seasonal variations has been
referred to as ‘seasonal forcing’ (Rohani, Earn, & Grenfell, 1999) and remains an
active field of exploration. Here we review evidence supporting the seasonal behav-
iour of infectious diseases. We focus on viral diseases and the ravaging coronavirus
disease 2019 (COVID-19).
COVID-19, and the fatal spread of its novel culprit, the severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2) pathogen, has emerged as the largest pan-
demic of the 21st century thus far (Wang, Horby, Hayden, & Gao, 2020). The first
case was reported in the Chinese city of Wuhan in December 2019 (Wu, Zhao, et al.,
2020). Since then the virus has spread over the world with more than 217 million
reported cases and 4.5 million deaths worldwide despite administration of over
5 billion vaccine doses as of September 1, 2021. Coronaviruses are a highly diverse
and globally distributed group of enveloped viruses with single-stranded RNA
genomes. They can infect humans and other mammals and avian species causing
Methods in Microbiology, Volume 50, ISSN 0580-9517, https://doi.org/10.1016/bs.mim.2021.10.002
Copyright © 2022 Elsevier Ltd. All rights reserved.
27
28 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
respiratory, gastrointestinal, hepatic, and neurological diseases. They are part of the
Coronaviridae family, subfamily Orthocoronaviridae, which consists of four coro-
navirus genera (α, β, γ, and δ) (Coronaviridae Study Group, 2020). To date, seven
human coronaviruses (HCoVs) have been identified as highly circulating viruses:
the α-coronaviruses HCoVs-NL63 and HCoVs-229E and the β-coronaviruses
HCoVs-OC43, HCoVs-HKU1, which cause seasonal and usually mild respiratory tract
infections, and the β-coronaviruses MERS-CoV, SARS-CoV-1 and SARS-CoV-2,
which cause severe and life-threatening respiratory disease (Ashour, Elkhatib,
Rahman, & Elshabrawy, 2020). SARS-CoV-2 infects humans and animals across dif-
ferent regions of the world. Many studies have been devoted to the causes of the
COVID-19 outbreak, its geographic distribution, factors that modify virus infectivity,
the effects of seasonal changes on viral transmissibility, and the genomic makeup of the
virus. Like many other respiratory diseases, COVID-19 shows an anticipated connec-
tion between climate and disease dynamics (Smit et al., 2020). It is therefore important
to study the seasonal behaviour of the SARS-CoV-2 virus and host responses that are
associated with infection since this knowledge can help mitigation efforts.
FIG. 1
The seasonal behaviour of measles and influenza. (A) The plot shows number of cases of
measles in Glasgow from 1900 to 1927. A noisy succession of biennial and annual cycles can
be observed spanning three periods. (B) Composite influenza virus activity in Chengdu, a
populous subtropical city in Southwestern China, from 2006 to 2015. Monthly positive rates
(in relative scale) were dissected into influenza A subtypes and influenza B lineages
(not shown). The start of H1N1 types of the 2019 pandemic and noisy semiannual and annual
cycles are indicated.
Panel (A) data from Soper, H.E. (1929). The interpretation of periodicity in disease prevalence. Journal
of the Royal Statistical Society, 92(1), 34–73. Panel (B) data from Zhou, L., Yang, H., Kuang, Y., Li, T.,
Xu, J., Li, S., et al. (2019). Temporal patterns of influenza A subtypes and B lineages across age in a subtropical
city, during pre-pandemic, pandemic, and postpandemic seasons. BMC Infectious Diseases, 19, 89.
two classical acute respiratory diseases caused by RNA viruses with significantly
different initial R reproduction metrics (12–18 and 1–2, respectively). While season-
ality appears not significantly constrained by infection spread potential (number of
infections caused by an infected person), the recurrence and stochastic behaviours
showcased in Fig. 1 are suggestive of the many research challenges that face the
study of seasonal behaviour.
A number of seasonal cycles of viral and bacterial diseases have been recently
documented in concert with their likely drivers (Martinez, 2018). Their ubiquity sug-
gests seasonality may be a unifying feature of epidemics in general. In some cases
there are clear cycles that impose temporal restrictions to infection despite vaccina-
tions or other mitigation strategies. For example, measles has been long associated to
school attendance, which fosters aggregation of school children and virus transmis-
sion (Fine & Clarkson, 1982; Soper, 1929). However, in some countries, measles
has been also associated to agricultural cycles (Duncan, Duncan, & Scott, 1997)
and the likely culprits of measles seasonality appear much more complex than
anticipated (Conlan & Grenfell, 2007). In fact, human physiology may be also an
important player.
30 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Seasonal variations play a vital role in determining the time at which an infection
might occur, its transmission, and its potential to become an epidemic (Rohani et al.,
1999). Important elements affecting seasonal cyclicity include biotic and abiotic
mechanisms. Seasonal cycles are sometimes associated with pathogen life cycles
that involve viral spread via insect vectors or maintenance in animal or environ-
mental reservoirs. To illustrate, MERS-CoV is recurrently introduced into the
Middle Eastern population during the camel calving season (Dudas, Carvalho,
Rambaut, & Bedford, 2018). Seasonal peaks of the Marburg virus disease coincide
with the birthing season of bats, which occurs twice a year (Amman et al., 2012).
Similarly, cases of rabies coincide with seasonal infection cycles in bats (George
et al., 2011). In zoonotic diseases such as these, seasonality may result from contact
with wildlife or livestock. In the case of Ebola, for example, the pathogen presence in
wildlife peaks in the dry season showing also an environmental effect (Groseth,
Feldmann, & Strong, 2007). Complex environmental and life cycle interactions also
occur in African sleeping sickness. The distribution of the tsetse fly vector, which
expands during the rainy season, affects the seasonal behaviour of the disease
(Knight, 1971). Seasonal changes in the incidence of vector-borne diseases such
as yellow fever, Zika and Lyme disease are also expected to be driven by seasonal
fluctuations in the population of the tick and mosquito (Aedes aegyptii) vectors (e.g.
Ferguson et al., 2016; Soper, 1967). This abiotic modulation of vector population
dynamics adds complexity to biotic mechanisms of seasonality. Abiotic mechanisms
are also central for some seasonal cycles. Factors include geographic location and
environmental conditions. For example, latitude influences the time of emergence
and the magnitude of disease outbreaks, such as in poliomyelitis (Paccaud, 1979)
and influenza (Cox & Fukuda, 1998). At worldwide level, early suggestions that in-
fluenza outbreaks move across the Earth every year along a sinuous curve parallel
with the ‘midsummer’ curve of vertical solar radiation (Hope-Simpson, 1981) have
been recently supported by an exhaustive modelling study (Deyle, Maher,
Hernandez, Basu, & Sugihara, 2016). Primarily, the seasons arise from Earth’s tilted
axis relative to the plane of its orbit, which changes the amount of sunlight reaching
its surface at various latitudes over the course of a year. Worldwide analysis and
modelling of the effects of meteorological factors of humidity and temperature sug-
gest periodicity of influenza follows seasonal periodicities of the planet but also
show a U-shaped nonlinear effect of absolute humidity on influenza, which is me-
diated by temperature (Deyle et al., 2016). This global effect produces a trade-off
of wetter air and low temperature that likely promotes viral spread by protecting
the proteins and lipids of the viral envelope. This trade-off appears optimal at
24 °C. Results align with previous experiments that used guinea pig models of
transmission to demonstrate the coordinated effects of temperature and humidity
on the incidence of influenza disease (Lowen, Mubareka, Steel, & Palese, 2007).
A number of environmental factors imposed by climate conditions are also expected
to play critical roles in epidemics as they influence pathogen survival during transi-
tion periods spanning hosts. These factors directly impact the survival of the virus
pathogen. These effects are particularly significant for respiratory viral diseases
2 The seasonal behaviour of viruses 31
(Moriyama, Hugentobler, & Iwasaki, 2020). For example, short exposure of viruses
to temperature or UV light exposure in droplets or other forms of fomite airborne
transmission can have significant effects on viral load and emergence of epidemics
(e.g. for influenza; Weber & Stilianakis, 2008). Alternatively, the environment can
also affect host susceptibility to infection via physiology or behaviour. Seasonal
variations are expected to be impacted by phenological effects of life history, includ-
ing cycles of migration or hibernation, and endogenous rhythm responsible for
seasonal changes in immunity, reproduction and metabolism, all of which are im-
pacted by the environment (Dowell, 2001; Martinez, 2018). Environmental factors
also affect antiviral defenses including intrinsic barriers such as mucus production
and epithelial integrity, inducible innate immune defense mechanisms, and adaptive
immunity (Moriyama et al., 2020).
FIG. 2
A triangle diagram of viral persistence explains main drivers of seasonality as a landscape of
trade-offs between factors related to environment, physiology and behaviour. The sphere
in the middle represents viral quasispecies clouds in Pareto fronts within a three-dimensional
fitness landscape. Viral interaction with environment, physiology and behaviour result in
approaches that achieve solution goals to increase persistence of viral life cycles.
(Nasir et al., 2017) explains how the three viral strategies of dependency (via
symbiotic-like associations of dissimilar entities tending towards mutualism, com-
mensalism, amensalism and parasitism), propagation (through lysis, budding and
transport), and dormancy (covert existence through latency via episomes or endo-
genized genetic material) take advantage of the engineering strategies of economy,
flexibility and robustness, respectively. Symbiotic-like co-dependencies impose altru-
istic, cooperative or antagonistic relationships that involve the budgeting of resources
(economy). Efficient propagation through lytic interactions requires establishing life
2 The seasonal behaviour of viruses 33
cycles and evolutionary arm races that foster co-evolutionary flexibility. Dormancy
inside cells shields against environmental variations and is a form of robustness.
The sphere in Fig. 2 represents a cloud of points in the trade-off space (morpho-
space) of a ‘viral quasispecies’, a dynamical virus collective defining a multiplicity
of virions and virocells (see Glossary). These points locate in Pareto fronts, which are
boundaries in multidimensional performance spaces that provide optimal fitness
solutions. Performance spaces are here defined as worlds of traits, which in our case
associate with strategies of economy, flexibility and robustness. Traits are the results
of processes such as number of cases in an epidemic, a geographical distribution of
vectors, or a direct measurement of seasonal behaviour (e.g. amplitudes of physio-
logical rhythms). These processes are sometimes drivers (e.g. schooling in measles
epidemics). The geometries of fitness solutions, which have been mathematically
elaborated (Sheftel, Shoval, Mayo, & Alon, 2013), correspond to planes in three-
dimensional performance spaces as described in Table 1. However, geometries
can be simpler, such as line segments arising from trade-offs between only two
drivers of strategy, or more complex, such as polygons describing trade-offs between
a multiplicity of drivers. Mathematical solutions to the analysis of performance
spaces is well developed and can provide new tools for the exploration of seasonality.
To explain drivers of seasonality, the triangle of viral persistence links environ-
ment to dormancy and robustness, physiology to propagation and flexibility, and
behaviour to dependency and economy (Table 1). These epidemic calendar associ-
ations become clear when seasonal drivers are assigned to strategies:
drivers, as has been made evident in the current socioeconomical and political
landscape of COVID-19. The fact that school cycles are linked to the seasonal
incidence of measles (Fine & Clarkson, 1982; Soper, 1929) comes as no
surprise. The same goes for other respiratory illnesses (e.g. varicella,
influenza). Many cultural, socioeconomic, and lifestyle factors affect seasonal
patterns of infection. Some of these factors change the frequency of interactions
between/within hosts, vectors and reservoirs and finally the host-virus
interface. Because human activity has been a dominant influence on climate
and the environment during the past centuries, anthropogenic influences are
expected to impact epidemic calendars. These include changing climate, land
and freshwater use, food availability and consumption, demographics, travel,
settlement and urbanization, technology developments, war and famine,
antimicrobial drug use, and even breakdown of public health measures (e.g.
Jones et al., 2008; Kronfeld-Schor et al., 2021). They also include effects
on geography, such as river fragmentations and dewatering (Farah-Perez,
Umaña-Villalobos, Picado-Barboza, & Anderson, 2020). All these effects can
be seen as dependency relationships that change behaviour at the level of the
organism and in doing so can impact epidemic cycles.
describe the structure of the data and its density function. For example, the scatter
plots of Fig. 3 show the relationship between the number of COVID-19 cases and
temperature, latitude or altitude of countries with reported cases. It is part of a larger
ongoing study (Hernandez and Caetano-Anolles, ms. in preparation) jumpstarted by
an initial exploration during the first wave of the pandemic (Burra et al., 2021). The
strong association of COVID-19 cases with temperature and latitude and its absence
with elevation suggests the geographical coordinate is associated with the onset
of the disease. Remarkably, the bimodal density patterns reveal countries located
at latitudes spanning 30–60°N or S and with average temperatures of 10–20 °C have
been most impacted. Bimodality was also present in Kernel density plots describing
the strong negative correlations between latitude and temperature (r ¼ –0.749;
rho ¼ –0.709), suggesting bimodality is at least partially driven by planetary environ-
mental factors. As we will describe below, these observations are relevant.
We note that correlations must first be confirmed as real, then every possible
causative relationship must be systematically explored. Five criteria must be fulfilled
to validate a causal relationship in scientific inquiry (Chambliss & Schutt, 2012):
(i) Empirical association: Typically this involves finding a correlation between
dependent and independent variables to determine if they vary together (covaria-
tion); without an empirical association of the variables there cannot be a causal
effect; (ii) Temporal priority: This criterion requires that there should be variation
in the independent variable before variation in the dependent variable, i.e. cause must
come before its effect; (iii) Non-spuriousness: Here the goal is to eliminate falsehood
in explanation of empirical associations by identifying other variables that could
explain empirical associations and subjecting them to a process of falsification;
(iv) Mechanism: Understanding a mechanism that connects independent and
dependent variables adds further support to the cause-and effect phenomenon; and
finally (v) Context: Establishing the external environment in which the cause-
and-effect occurs, for example, with networks of cause-effect relationships, the
causal-effect relationships are strengthened.
A number of tools help explore causal relationships, such as using hypothesis
testing methods that compare null and auxiliary hypotheses to a primary hypothesis,
or using simple controlled experiments (e.g. split-run testing in randomized exper-
iments) that compare ad minimum two versions of a single variable. Here, Fisher’s
statistical principles of experimental design—comparison, replication, randomiza-
tion, blocking, orthogonality, and factorials—are helpful. They seeded advances
in algebra and combinatorics and development of impactful methods such as the
Taguchi methods for robust engineering design that for example transformed the
automotive industry (Montgomery, 2013).
season peaks somewhere between November and March. Besides stochastic behav-
iour, there appears to be higher-level recurrent events imposed on the typical cycles
in measles and the flu that must be explained, such as semiannual or biennial behav-
iour. The existence of pandemic or major epidemic events complicate the recurrent
patterns, as showcased by the incidence of influenza A and B in a major metropolis
of Northwestern China following the 2009 H1N1 pandemic (Zhou et al., 2019;
Fig. 1B). Similar complications have been also highlighted for the city of Mexico
(Bjørnstad & Viboud, 2016).
Statistical predictive and modelling methodologies must be developed when
principles driving a dynamical system are poorly understood. Such is the case of
the seasonal behaviour of viruses. Time series analysis is a methodology readily used
in public health and biomedicine applications (Zeger, Irizarry, & Peng, 2006). It
addresses experimental situations where a sequence of observations is made over
time to monitor equally spaced temporal changes of a phenomenon, such as the
number of cases of viral disease in a population. The goal of the methodology is
to generate simple descriptions, explanations, predictions or control of the underly-
ing process. The plots of Fig. 1 for example are simple descriptions of a temporal
recurrence. Data displays and summary statistics are created to better understand re-
sponse variations over time. In some cases, the time series can be decomposed into
components to reveal autocorrelations that can help us better describe the process
(e.g. dissecting trends, seasonality, and residual effects). Sometimes a time series
can be decomposed into a numerous series of components, using the Fourier trans-
form or the discrete wavelet transform, for example. The time series is often
re-expressed as cosine waves of arbitrary amplitude and phase. Amplitudes can
be plotted against frequency to produce ‘periodograms’, and a ‘spectrum’ of the sto-
chastic process can be estimated as the expected squared amplitude. When the goal of
statistical analysis is explanation, the dependence of the process is modelled against
one or more predictor time series using regression analysis. These predictor series are
assumed non-random. Because neighbouring values of the process tend to be corre-
lated, we can make predictions from past process responses or from predictor covar-
iate time series with autoregressive models or distributed lag models. Time series
regression models conditioned on past observations can take the form of marginal
or conditional models, which often dissect a regression model for the mean and a
model for the autocorrelation function. In the presence of Gaussian processes, auto-
regressive moving-average models display autocorrelation functions that decay or
oscillate to zero with increasing lag. In some cases, the autoregressive process ex-
hibits error and the models used to describe it are stationary and do not exhibit
long-term trends and are therefore only appropriate in the absence of trends. Simi-
larly, in cases where past responses influence the present through non-linear func-
tions, we must apply nonlinear time series modelling approaches that for example
can take into consideration response interactions. Finally, one popular approach
to dissect the noisy time-series data that embody seasonal disease cyclicity, is the
use of nonparametric autoregression methods (Hardle, 1990), including nearest-
neighbours, splines, local polynomials and neural nets. These methods recursively
40 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
dissect error from chaotic behaviour that may govern cyclic disease patterns in
biology (Sugihara, Grenfell, & May, 1990). For processes underlying periodic
behaviour, a general form of spectral density may be assumed with increasing sample
size that can dissect the autocorrelation structure of the stationary process (Hardle,
L€utkenpohl, & Cheng, 1997). A recent analysis of global environmental drivers of
influenza showcases the power of using nonparametric autoregression in the form
of a smoothing spline to determine seasonal cycles (Deyle et al., 2016). The spline
works on time series variables with and without sinusoidal waveforms.
FIG. 4 The seasonal behaviour of COVID-19. (A) The plot shows the numbers of confirmed
cases (black) and weekly deaths (red) since the start of the recorded COVID-19 pandemic.
Three waves are clearly evident for worldwide weekly deaths recorded at the time of data
collection from CSSE at John Hopkins University, September 1, 2021. Data gathered in April
15, 2020 (Burra et al., 2021) (a) and May 23, 2021 (Hernandez and Caetano-Anolles, ms. in
preparation) (b) are highlighted in the timeline and its analysis summarized in panel (D).
(B) Contour plot of temperatures taken 2 metres above Earth’s surface for the entire world
during the first wave of the pandemic (November 2019 to March 2020). Data was
extracted from the NCEP/NCAR reanalysis (v. 1) project and rendered online (Climate
Change Institute, University of Maine; climatereanalyzer.org). White circles represent
countries with substantial outbreaks (>10 cumulative confirmed deaths per million people)
as of April 15, 2020. With exception of Peru, Ecuador and the Dominican Republic, all country
outbreaks occurred in yellow or green-shaded contours with temperatures ranging 5–11 °C.
(C) A timeline of countries with substantial outbreaks (>10 cumulative confirmed deaths per
million people) showing COVID-19 spread massively in a 30°N to 60°N corridor of latitudes
during the first wave of the pandemic and then spread through a 30°S to 50°S corridor of
latitudes during the onset of the winter season of the Southern Hemisphere. Red circles are
countries exhibiting >300 confirmed deaths per million people as of November 15, 2020.
(D) Effect of temperature (Temp.) and geographical coordinates of Latitude (Lat.) and
Longitude (Long.) on number of cases and deaths worldwide normalized to account for
population differences of the 211 and 191 countries analysed in 2020 and 2021, respectively.
Correlations were considered significant when P-values were less than 0.05 and association
strengths had Pearson’s correlation coefficients r were higher than 0.1. (E) Seasonal
oscillations detected in 5 countries of the Northern Hemisphere (NH), USA, India, Russia,
France and the United Kingdom, and 5 countries of the Southern Hemisphere (SH), Brazil,
Argentina, South Africa, Peru and Chile. Average seasonal signals and 95% confidence
intervals are given in red and black shades for the two hemispheres, respectively.
Seasonal signals were detected in the time series of cases and deaths using an adaptive
one-dimensional time series analysis, the Ensemble Empirical Mode Decomposition
(EEMD) method.
Panel (E) data from Liu, X., Huang, J., Li, C., Zhao, Y., Wang, D., Huang, Z., et al. (2021). The role of
seasonality in the spread of COVID-19 pandemic. Environmental Research, 195, 110874.
3 The seasonal behaviour of COVID-19 43
convection, circulation and biomass. The Earth’s 23.5° tilted axis and its elliptical
orbit around the sun make radiation strike the surface at different angles following
the ecliptic of the celestial sphere. This causes differences in solar energy that create
cyclic variations in temperature at daily and yearly levels. We note that temperature
also varies with differences in altitude and topographical surface. For example, con-
tinents are generally warmer than oceanic regions in the Northern Hemisphere, while
this reverses in the Southern Hemisphere tempered by the scarcity of land masses. If
temperature variation impacts the triangle of viral persistence, a viral pandemic of
COVID-19 proportions is expected to leave increasingly stronger worldwide sea-
sonal signatures as the disease progresses. In addition, since distance from the Equa-
tor measured as latitude is a strong predictor of temperature, any temperature effects
on seasonal behaviour detected by studying correlations of geographic coordinates
with epidemiological data should be strengthened. The same can be said with other
variables, such as elevation, which is correlated with UV radiation.
Indeed, initial studies during the first wave of the pandemic (Fig. 4A) suggested
temperature and humidity levels of geographical areas (countries, regions, cities)
were inversely related to epidemiological variables such as confirmed cases or
deaths, while latitude showed a positive relationship. In many instances however re-
sults were inconclusive. A GRADE tool analysis of 17 initial explorations (mostly
non-peer reviewed at that time) of data downloaded prior to March 1, 2020 (generally
much earlier) revealed that only 4 were low risk (Mecenas, Bastos, Vallinoto, &
Normando, 2020). Of these four, the only formally published study concluded that
weather factors alone could not explain variability of the viral reproductive number
in Chinese provinces or cities (Poirier et al., 2020). The meta-analysis suggested
warm and wet climates reduced COVID-19 spread but also highlighted the impor-
tance of controlling confounding variables. Other initial studies of epidemiological
variables were of significance. Demongeot, Flet-Berliac, and Seligmann (2020)
showed that higher temperatures decreased infection rates in both French adminis-
trative regions and in 21 countries spread throughout continents. An association anal-
ysis of temperatures of countries or regions with epidemiological variables collected
at the beginning of March 2020 and a time series predictive analysis with the Auto-
regressive Integrated Moving Average (ARIMA) model showed COVID-19 speed of
contagion decreased with increasing temperatures. In addition, a focused study of
how weather parameters of temperature, humidity and rainfall affected disease inci-
dence ending March 2020 in Jakarta, Indonesia, revealed only average temperature
exhibited statistically significant associations (Tosepu et al., 2020). The use of a log-
linear generalized additive model to study cases and deaths in 166 countries with
March 27, 2020 data showed that a temperature increase of only 1 °C reduced the
number of daily reported cases by 3.08%, while a 1% increase in relative humidity
reduced the number of daily reported cases by 0.85% (Wu, Jing, et al., 2020). The
analysis concluded high temperature and humidity significantly affected COVID-19
epidemiological variables. Similarly, data from 429 cities also showed a negative
correlation between viral transmission and temperature, reporting that a 1 °C temper-
ature increase reduced the reported cases by 0.86% (Wang, Jiang, et al., 2020).
44 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Temperature, population age, and tourism were the most important factors affecting
COVID-19 risk measured with cases and deaths in 154 countries at the end of March
2020 (Yang & Lee, 2021). The study used a Pearson’s correlation matrix but also
modelled nonlinear associations of temperature and risk with random forest statisti-
cal methods. In contrast, Kassem (2020) reported the impact of temperature on cases
per million in 43 countries divided into three groups according to first time disease
introduction. There was only a slightly significant (P < 0.1) inverse relationship in
only one of the groups.
While temperature and humidity were the initial focus, an analysis of 50 cities
worldwide with and without community spread as of March 10, 2020 showed com-
munity transmission was restricted to a 30°N to 50°N latitude corridor with consis-
tent weather patterns of 5–11 °C average temperatures combined with low absolute
humidity (Sajadi et al., 2020). Poole (2020) also observed a similar corridor spanning
25°N to 55°N latitudes. Indeed, a world temperature map showed countries with sig-
nificant COVID-19 outbreaks during the first wave of the pandemic (exhibiting
>10 cumulative deaths per million people as of April 15, 2020) were located within
a 30°N to 60°N latitude corridor in regions with 5–11 °C average surface tempera-
tures (Fig. 4B). Moreover, a timeline of countries with significant outbreaks
(>10 cumulative deaths per million people) describing how COVID-19 was advanc-
ing from China to the rest of the world for the most part of year 2020 shows that the
disease began spreading through a latitude corridor in the Northern Hemisphere
followed by one in the Southern Hemisphere (Fig. 4C). Almost half of outbreak
countries were in the Northern corridor, 40% of which showed >300 cumulative
deaths per million people as of November 15, 2020. A transition from the Northern
corridor to Southern latitudes began to occur at the beginning of April with outbreaks
in Peru and Ecuador (when temperatures started to lower in the Andean mountain
range), then Brazil (beginning with the elevated Sao Paulo) but reached the Southern
corridor in May and fully in June (when winter formally started in the Southern
hemisphere). We also explored the worldwide role of temperature and geographical
coordinates of latitude and longitude on epidemiological parameters for 211 coun-
tries at the peak of the first wave, April 15, 2020 (Burra et al., 2021). Both temper-
ature and latitude were significantly correlated to COVID-19 cases and deaths
with weak to moderate association values (Fig. 4D). Repeating the analysis during
the third wave, a year later, showed these associations were becoming stronger
(Figs 3 and 4D). This corroborates the impact of the geographical temperature-
latitude correlate on COVID-19 seasonal behaviour, which is likely associated with
the planetary effect of solar radiation. Thus, latitude, temperature and humidity
appeared important factors during the first wave of the pandemic, consistent with
the behaviour of a seasonal respiratory virus.
Given the strong temperature-latitude association we detected, COVID-19 sea-
sonality is likely related to planetary-level environmental effects of solar radiation
on viral transmission, directly through germicidal effects or indirectly through ‘phys-
iology’ effects (vitamin D-related or through other immune regulatory activities).
The low-energy infrared (IR) wavelengths transmit heat and could be responsible
3 The seasonal behaviour of COVID-19 45
for germicidal effects of temperature. Laboratory studies have shown that high tem-
peratures and humidity levels shorten SARS-CoV-2 half-life in aerosols and other
media, with virus persistence dramatically decreasing with small temperature in-
creases (Chin et al., 2020; Matson et al., 2020; van Doremalen et al., 2020). The
high-energy ultraviolet (UV) wavelengths (especially the UV-C spectrum) could
have direct germicidal action on viral survival, as shown in laboratory experiments
(Heilingloh et al., 2020; Seyer & Sanlidag, 2020). The expectation that UV radiation
levels could be correlated with COVID-19 cases and deaths was recently fulfilled
(Karapiperis et al., 2021). A machine learning-based analysis of data from 12 coun-
tries distributed across latitudes with comparable economic and demographic indices
and epidemic surveillance statistics showed year-round UV radiation levels strongly
associated with incidence rates, more so than with other variables. Remarkably, time
series showed high UV levels depressed the number of cases in a hemisphere-related
manner. These types of studies link geographical to temporal effects of seasonality
and may lift some misconceptions about weather and seasonality (Carlson, Gomez,
Bansal, & Ryan, 2020) by providing new methods of hypothesis testing.
in iterative steps allowed to obtain ensemble means of the mode functions for signal
extraction. Remarkably, the signal curves showed COVID-19 cases and deaths were
higher in colder climates and seasonality more pronounced at higher latitudes in an
hemisphere-related sinusoidal pattern (Fig. 4E). This provides further confirmation
of the seasonal behaviour of COVID-19.
Remarkably, these low PID levels are not only a rigidity property of coronaviruses
alone but of the entire Nidovirales group. A global analysis of 6044 viral proteomes
showed levels of mean intrinsic disorder were less than 0.2 for the proteomes of the
Nidovirales (Fig. 5C).
In Section 4, we focus on possible molecular culprits of seasonal behaviour. We
study the variants of the SARS-CoV-2 proteome encoded in thousands of genomes
sampled during the first wave of the pandemic. Analysis reveals that a number of
molecular regions associated with clades and the highly infectious ‘variants of con-
cern’ (VOCs; see Glossary) that arose in later waves are located in PID regions we
initially identified that are necessary to sustain the structural integrity of virion struc-
ture. We also show that the N-terminal domain of the S-protein, which decorates the
outer shell of the virion particle (crowning its ‘corona’ appearance), contains a
galectin-like structure. Tracking the prevalence of mutations in this structure follows
a seasonal pattern. We propose the galectin-like structure is a frequent target of
mutations because it helps the virus evade or modulate physiological responses of
the host to further its spread and survival. We end by suggesting that these regions
are good molecular candidates for sensors of environment and physiology.
deleterious and hinder their viability (Johnson & Barton, 2002). This puts RNA vi-
ruses close to an ‘error threshold’ of too many deleterious mutations for successful
persistence (Domingo, Sheldon, & Perales, 2012). Remarkably, coronavirus proof-
reading abilities are crucially enhanced by the 30 -to-50 exonuclease domain of the
NSP14 protein (Minskaia et al., 2006), which increases 12- to 15-fold template copy-
ing fidelity (Eckerle et al., 2010) and enables both genome expansion and mutation
robustness (Smith & Denison, 2013). Genetic recombination and genomic reassort-
ment also contribute to viral diversity and emergence of new phenotypes. In recom-
bination, two or more segments from two different parental genomic molecules
combine and produce a new mutant or recombinant phenotype that may be more
pathogenic and virulent than the parental molecules (Posada, Crandall, & Holmes,
2002). Recombination may accelerate viral evolution, but more research is needed
to evaluate the exact role it plays (Rhodes, Wargo, & Hu, 2003). Genomic reassort-
ment occurs in tripartite genomic RNA viruses like influenza in which new variants
arise after the simultaneous entry of two or more different viruses into a host cell
(Vijaykrishna, Mujerki, & Smith, 2015). Recombinants produced by genetic recom-
bination exhibit new properties that can significantly enhance viral diversity (Earn,
Dushoff, & Levin, 2002). In fact, the high recombination rates of RNA viruses are
directly associated with their virulence (Khatchikian, Orlich, & Rott, 1989), the host
range (Gibbs & Weiller, 1999), host immunity (Malim & Emerman, 2001), and
resistance to antivirals (Nora et al., 2007).
While mutation frequency determines genetic variations in a viral population and
speeds up viral evolution (Sanjuán & Domingo-Calap, 2016), mutational robustness
is dependent on population size and how viruses manage the detrimental effects of
mutations. While most random mutations (approximately 40%) are lethal, deleteri-
ous mutations that are non-lethal (approximately 30%) lead to a reduced effective-
ness of the virus (Sanjuán, Moya, & Elena, 2004) or can have positive effects, as has
been reported in bacteriophage φ6 (Burch & Chao, 2004), vesicular stomatitis virus
(Sanjuán et al., 2004), and HIV-1 (Bonhoeffer, Chappey, Parkin, Whitcomb, &
Petropoulos, 2004). However, beneficial mutations may occur if the environment
changes rapidly, if a multiplicity of ecological niches are present, or if other factors
adding to the viral mutational landscape enhances novelty generation, including rear-
rangement/recombination, changes in genome size, host-encoded modifiers, and rep-
lication mode (Sanjuán & Domingo-Calap, 2016). Regardless of how viruses
manage mutations, changes in population size can influence the frequency of a given
mutation and the identity of the viral population as a whole. For example, small
populations let genetic drift cause stochastic fixation of mutations in sequences that
are conserved and are present in the parental genome. Because most of the progeny
will acquire these mutations, the identity of the population is maintained, very much
as it occurs in higher organisms. In other words, a small population size guarantees
‘survival of the fittest’ by competition and natural selection of the best-adapted var-
iants among a limited number of genomes (Earn et al., 2002). When populations are
large, a swarm of genetic variants revolving around a consensus sequence is formed
in a phenomenon known as ‘the survival of the flattest’ (Lauring & Andino, 2010).
4 Viral genomic make up and seasonality 51
Here, viruses with larger numbers of variants are retained, limited only by the ‘error
threshold’ and the catastrophic possibility of viral extinction. Viral identity is lost
and replaced by a ‘quasispecies’, a group of different variants that show genetic
linkage through mutation, have shared functions, and collectively contribute to
the characteristics of the population.
(suggesting fixation) and its value increases when mutations are diluted in the viral
cloud. The second variable is a relative measure of entropy, ‘relative entropy delta’,
which measures selection pressure by comparing viral diversity at two different time
points of a time series. Entropy reversal trends suggest mutations are positively
selected to enhance fitness while entropy expansions suggest other evolutionary
forces are at play. We note entropy expansions are expected to precede reversals
since they generate novelties which are then culled in a reversal phase.
Out of the 27 high-entropy mutations identified, 19 were entropy reversals
(Table 2). Five of these have been confirmed as positively selected and exhibiting
population level expansions (as of March 2021; Singh et al., 2021). Another 4 pos-
itively selected mutations were associated with the PID-rich serine/arginine-rich
linker of the N-protein, R203K and G204R, and the viroporin protein encoded by
ORF3a, Q57H and G251V. They were in either entropic expansion or reversal
modes, respectively. Most of 27 high-entropy mutations were located on the surface
of the molecules once mutations were traced onto three-dimensional atomic models
(Tomaszewski et al., 2020). The only exceptions were the R203K and G204R mu-
tations of the N-protein, which were buried, and the G57H mutation of the ORF3a
viroporin, which was located on an internal transmembrane domain. Remarkably,
seven of the high-entropy mutations were located on PID regions of significant dis-
order of the N-protein, the NSP3 papain-like protease, and the ORF3a viroporin
(Table 2). Three of these were positively selected.
Our analysis identified the well-known D614G mutation of the S-protein had co-
ordinated entropic trends with the P323L mutation of the NSP12 polymerase that
mediates viral replication. The D614G mutation, which is already fixed worldwide
in the current viral population, was part of a haplotype of four mutations altering the
S and NSP12 proteins, 50 UTR, and silently the NSP3 papain-like protein (F106F)
during the first wave. This haplotype defined the G-clade that originated in China
and was established in Europe in the middle of January 2020 following its first report
in Germany (Korber et al., 2020; Phan, 2020). A time series of variant appearance
revealed that the D614G and P323L haplotype marker of the G-clade had spread
through continents during the January-April period of the first wave visualized
through genomic analysis of the thousands of sequences we had available at the time.
The haplotype is believed to have increased infectivity (Becerra-Flores & Cardozo,
2020; Korber et al., 2020), and as previously discussed, to have enhanced protein
flexibility (Korber et al., 2020).
We also revealed accumulation of high-entropy variants enhancing flexibility in
PID regions and other regions that were of interest. The N-protein plays critical roles
in maintaining viral structure and viability once the virus enters the cell, helping viral
replication, transcription and packaging (e.g. Woo, Lee, Lee, Kim, & Cho, 2019).
The N-proteins holds two major RNA-binding domains, an N-terminal domain
(NTD) and a C-terminal domain (CTD) connected by a central serine/arginine-rich
linker and flanked by terminal sequences. We found these linker and terminal
sequences were PID regions and their contribution to the entire molecule made
56 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
immunodominant (i.e. is the preferred epitope), changes in the NTD region alter
antigenicity (reviewed by Harvey et al., 2021). In fact, an ‘NTD supersite’ consisting
of NTD residues 14–20, 140–158 and 245–264 is involved in antibody neutralization
by inhibiting conformational changes or interactions with auxiliary receptors of its
structurally plastic structure. NTD is also central from an evolutionary perspective.
An analysis of ‘evolutionarily important’ residues of the S-protein detected by
sequence conservation showed they were concentrated in two domains, the NTD
and the RBD, both of which had a role of host cell binding in a number of related
viruses (Saputri et al., 2020). While many evolutionarily important residues were
detected in NTD, suggesting an involvement in host receptor binding, the study
showed ‘importance’ correlated with structural flexibility. Remarkably, all atom
molecular dynamics simulations of the SARS-CoV-2 S-protein revealed protein
flexibility was dependent on external temperature (Rath & Kumar, 2020). In the sim-
ulations, the RBD was less mobile than the NTD, with flexibility limited mostly to
the receptor binding motif. Increasing temperature made a number of charged polar
amino acid residues on the top layer of the more flexible NTD less solvent exposed.
These residues involved loops delimited by the N-terminal β-strand, β8-β9, β9-β10
and β14-β15, which harbour most mutations highlighted in Fig. 7D (including the
inset). Similarly, increases in temperature closed the flexible binding motifs of
the RBD in the trimer, which are usually in an open conformation. This conformation
sealed the visibility of the trimeric pore when visualized from the top of the spike,
burying the receptor binding residues necessary for contacting the ACE2 receptor.
Thus, temperatures above 40 °C resulted in the inactivation of the spike, possibly
in a reversible manner and without loss of secondary structure. The combination
of mobile rigid elements encompassed by deformable regions that act as hinges
may be the mechanism behind this environmental modulation. Given these results,
the putative binding properties of NTD are likely to have a regulatory role related to
environmental responses, which we here sought to confirm.
Finally, the significant appearance in clade GRY of the N501Y mutation (Fig. 6)
revealed the early accumulation of a marker in one of the six key contact residues of
RBD shown to increase both ACE2 receptor affinity and infectivity and virulence
(Starr et al., 2020). A cryo-EM analysis of receptor-triggered dynamic transforma-
tion from the closed prefusion state of the spike to the fusion-prone open state
(Fig. 7C) exhibited enhanced sensitivity of ACE2 through the T470-478 loop and
Y505 site of RDB viral determinants (Xu et al., 2021). Remarkably, T478K has
become a significant marker of VOC delta. The appearance of VOC alpha at the
end of 2020 brought also the P681H marker of the S-protein, one of four residues
comprising the insertion that creates a S1/S2 furin cleavage site between S1 and
S2 in spike, which is known to promote viral entry and transmission in animal
models. Harvey et al. (2021) carefully reviewed how mutations associated with
the main binding function of the S-protein and other functional regions that appeared
in this later phase of the pandemic contributed to ‘immune escape’ by changing an-
tigenicity and transmissibility but also reducing the efficiency of vaccines, convales-
cent plasma, and post vaccination serum. None of these markers were significant in
60 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
our initial exploration of high-entropy mutations during the first wave of the
pandemic, showing they represented novel targets of dynamic change in the viral
population.
Thus, the three structuring phases proposed in Fig. 6, ‘flexibility/rigidity’, ‘envi-
ronmental sensing’, and ‘immune escape’, are compatible with the gradual appear-
ance of markers over time associated with multiple PID regions, a putative molecular
sensor located in evolutionarily important sites of the N-terminal NTD region of the
S-protein, and the central receptor-binding function of the RBD region, respectively.
[NTD plus signal peptide (SP)], the RBD region, and in ‘Other’ regions, including
the S2 subdomain of the S-protein. The time series showed the early and growing
prevalence of the D614G variant (90% during the week of June 19) but also of
the NTD L18F and A222V substitutions, which spread in July and early August,
respectively. Most RBD substitutions peaked later. For example, S477N peaked
in July (with 44% prevalence worldwide and massively present in Oceania) but
quickly decreased in incidence. RBD substitution N439K and NTD deletion
H69_V70del were significant but were clearly late introductions.
More importantly, the timeline revealed a cyclic pattern of prevalence for the
N-terminal region, but not so much for RDB or ‘Other’ regions. With the exception
of D614G and SP variant L5F, which progressed towards fixation, all mutants intro-
duced during the January and February winter months of the Northern Hemisphere
showed significant bursts of genomic abundance lasting one or a few weeks, which
then declined significantly in later months with some resurfacing by the end of 2020.
These included the first 6 mutations in the N-terminal SP and NTD regions (H49Y,
S98F, Y28H, L8V and L5F), two mutations in the intracellular domain (ID)
(T1273extQ and P1263L), one in the cytoplasmic domain (CD) (W1214X) and
one in the RDB region (V367F). ID and RDB mutations later vanished as the year
progressed. With only one exception (D253G of the NTD), mutations appearing be-
tween the months of March and July had low prevalence and accumulated without
significant bursts in the timeline. No significant late mutation entries appeared after
July as the Northern Hemisphere was entering summer, again suggesting a seasonal
component.
Remarkably, dissecting the timeline along geography uncovered bursts occurring
preferably in the N-terminal SP and NTD regions that increased during the winter
seasons of the Northern Hemisphere for Europe, North America and Asia (Fig.
8B), and the winter season of the Southern Hemisphere for Oceania, South America
and in part Africa (Fig. 8C). This expected seasonal pattern, which matches patterns
shown in Fig. 4E, provides strong support to assumptions and hypotheses. The
hemisphere-related sinusoidal pattern was particularly evident in Asia and South
America. In Asia, a significant number of bursts in January-March driven by the start
of the pandemic in China and involving either the NTD or ‘Other’ regions of the
S-protein was followed by marked decreases and then significant increases in activ-
ity towards the end of the year with massive numbers of bursts involving the NTD
region. In South America, the pattern reverses with most bursts occurring during the
months of May and June, fewer bursts observed in July and August, and no burst
observed in September and October, with patterns mostly driven by variants in
the ‘N-terminal’ and ‘Other’ categories. A similar behaviour was observed for Africa
and Oceania but with one difference for the number of bursts during September and
October for Oceania, which was maximal for that set. We note that the absence of
entries during the months of January and February for South America and Africa may
be related to the low numbers of sequenced genomes available for those regions dur-
ing that time. We also note that all of North America and Europe and most of Asia
(with exception of East Timor, Indonesia, and Maldives) is located on the Northern
4 Viral genomic make up and seasonality 63
Hemisphere and that most of South America (90%), all of Oceania and only 30% of
Africa is located in the Southern Hemisphere, which holds only 15% of the world
population. For Africa, only the Mediterranean countries and South Africa are
within the seasonal corridor of the disease. Despite possible geographical uncer-
tainties, the hemisphere-related patterns appear consistent.
How would bursts exert their influence on seasonality? Quantifying variants pre-
sent in a sequence provides insight into the kind of changes occurring in an individual
burst, which depend on the functional needs of proteins at that time. We therefore ex-
plored the combinatorial landscape of variants arising during the 2020 timeline. Out of
137,605 variant sequences analysed (Showers et al., 2021), 9.8% contained only one
variant (D614G), while 44.7% contained 2, 30.8% contained 3, and 14.4% contained
4–9. The average of 2.53 0.94 (SE) variants per sequence shows the poor mutational
saturation of the sampled viral population as a whole. A network of variants with links
describing their co-occurrence in sequences revealed that the vast majority of
sequences had variants that co-occurred with D614G. Thus, D614G represents the
central hub of the network. In fact, only 43 variant combinations representing 128
sequences lacked D614G variants (3 of which however co-occurred with other variants
of the 614 site). This is surprising and suggests D614G plays a very central functional
role, possibly of flexibility, that is complemented by each new variant. A network of
co-occurrence of variants in sequences with variant combinations present in at least
500 sequences is shown in Fig. 8D. Remarkably, the network reveals that variants
located in the NTD region, especially A222V and L18F, were driving network
co-occurrence in 2020. This again supports the central role of NTD as environmental
sensor. Note that 7,088 sequences contained all three D614G, A222V and L18F var-
iants, the top most common 3-variant combination. As expected, many of the central
network players we mapped earlier onto the atomic model of the S-protein (Fig. 7)
were also central hubs in this highly represented network.
effects were not appropriately dissected in the genomic set. Alternatively, and given
the results of Fig. 8, variant bursts rather than variant levels could harbour better
signatures of seasonal behaviour. It is unclear whether the evolution of the SARS-
CoV-2 quasispecies is driven by mutational load and not specific molecular sensors,
as we here propose. Further exploration is therefore warranted.
FIG. 9 Galectins, likely culprits of COVID-19 seasonal behaviour. (A) The DALI structural
neighbourhood of the spike protein of SARS-CoV-2 (PDB entry 6vxxA) contains 2310
structures (downloaded June 3, 2020) spread through a scatter plot describing their RMSD
and Z-score values (see Glossary for definitions). RMSD describes rigid-body structural
superposition while the Z-score describes a length-rescaled distance matrix alignment that
maximizes one-to-one atomic correspondences between two structures with a weighted sum
of similarities of intramolecular distances. The plot reveals galectins (red circles) exhibit a
good structural match (RMSD 2.5–5 Å) to a small segment (Z-score 8–10) of the spike
protein query (which self-aligns with RMSD ¼ 0 and Z-score ¼ 50). The inset shows the
match correlation matrix, which describes the correlation of matched structures along amino
acid residue positions of the query structure (proceeding from N-terminus to the C-terminus
in the x axis). The matrix can be used to evaluate modular segments in the alignment of the
entire neighbourhood. Structural modular domains are expected to show strong positive
correlation (red hues) within domains and negative correlations (blue hues) between
domains. The highlighted white square at the N-terminus of the molecule in the left-bottom
part of the correlation matrix reveals that the region of the N-terminal domain (NTD) of domain
S1 of the spike, which is homologous to the galectin fold (see panel B) is indeed modular.
The matrix also shows significant structural modularity of the receptor binding domain (RDB)
and the S2 subunit of the spike molecule. (B) Structural alignment of the monomer of
galectin-4 (brown; PDB entry 3ap5A) to the N-terminal domain of the coronavirus protein
(green; PDB entry 6vxxA) shows there is a good structural match in their three-dimensional
atomic models (rendered in ribbon format), confirming structural similarities (putative
homologies) between the NTD and the galectin fold. Alignments with other galectins show
similar structural matches (not shown). (C) A protein sequence and secondary structure
alignment details the structural match at the N-terminal region of the spike molecule.
Symbols of amino acids in the pairwise sequence alignment between the query SARS-CoV-2
spike protein and galectin-4 at the top are given in the one-letter amino acid notation of the
IUPAC-IUB Commission of Biochemical Nomenclature. Symbols in the pairwise
alignment of secondary structures at the bottom follow DSSP nomenclature: H/h, helix; E/e,
strand; L/l, coil. Uppercase means structurally equivalent positions with the query sequence.
4 Viral genomic make up and seasonality 67
Z-score scatter plot detected a multiplicity of structural clusters (blue circles in the
plot), most of them related to spike proteins of other RNA viruses (Fig. 9A). While
these spike structures showed good structural matches (RMSD 2–5 Å), alignments
of protein segments of decreasing length (Z 10–50) showcased the remarkable
structural diversity that exist in the world of spike proteins. The novelty is the exis-
tence of a cluster of small structures with RMSD 2.5–5 Å and Z 8–10 (red cir-
cles) showing a small segment located on the N-terminal region of the SARS-CoV-2
spike protein (see correlation matrix in Fig. 9A) matched known crystallographic
structures of galectin proteins. Fig. 9B and C show structural and secondary structure
alignments between galectin-4 and the spike protein we used as query. Note align-
ment of the extended strands (E) that make the sequential loops of β-strands.
But why are galectin-like structures relevant to the seasonal behaviour of
COVID-19? The answer can be found in the coral reefs of the Pacific and Indian
Oceans and the devastating phenomenon of coral bleaching that threatens marine
resources and the well-being of the millions that live in coastal communities.
Scleractinian corals establish an endosymbiosis with dinoflagellates of the genus
Symbiodinium that allows them to grow and persist in warm, well-illuminated and
oligotrophic waters. Coral bleaching has been attributed to symbiosis disruption un-
der environmental stress resulting from rising ocean water temperatures and other
factors of global change (e.g. Kleypas, Castruccio, Curchitser, & McLeod, 2015).
A number of lectins have been identified in corals as part of the innate immune
response system of coral communities (Kvennefors et al., 2010), some implicated
in thermal and disease stress responses (Ricci et al., 2019; Vidal-Dupiol et al.,
2014). In particular, galectin proteins with antimicrobial immunity functions were
found in greater abundance in healthy coral symbioses (Ricci et al., 2019). Remark-
ably, a recent study showed galectin functions of the scleractinian coral Pocillopora
damicornis were dependent on temperature effects, establishing galectins as possible
environmental sensors (Wu et al., 2019). A cloned galectin that encoded a 293 amino
acid-long protein harbouring a CRD and collagen-like N-terminal tail with signal
peptide was able to recognize and agglutinate or stabilize coral pathogens and sym-
bionts. Binding activities were only optimal between 25–30 °C. Results suggested
galectins may be involved in heat bleaching of scleractinian corals through
temperature-regulated recognition of pathogens and symbionts.
We end by noting that lectins are a diverse family of proteins with the capacity to
selectively recognize carbohydrate moieties associated for example with receptor
proteins. It appears that the SARS-CoV-2 S-protein, besides binding ACE2, also
interacts with the CRD of C-type lectin receptors, which facilitate viral entry by
dysregulating host immune responses and acting as entry receptors (Rahimi,
2021). A number of studies have recently surfaced that point into the centrality of
lectin-mediated recognition (Gao et al., 2021; Soh et al., 2021; Thepaut et al.,
2021). Curiously, these lectins interact with both ACE2 and with the NTD domain
of the S-protein (Soh et al., 2021), perhaps facilitating the process of receptor
presentation by trans-infection (Thepaut et al., 2021).
68 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Acknowledgements
We dedicate this work to the frontline medical professionals who have been saving the life of
others with limited protective equipment, selflessly, and at their own peril during an age of
misinformation. COVID-19 research in the laboratory of G.C.-A. is supported by the Office
of Research and Office of International Programs in the College of Agricultural, Consumer
and Environmental Sciences at the University of Illinois at Urbana-Champaign.
References
Abotaleb, M., & Makarovskikh, T. (2021). System for forecasting COVID-19 cases using
time-series and neural networks models. Engineering Proceedings, 5, 46.
Akhila, M. V., Narwani, T. J., Floch, A., Maljkovic, M., Bisoo, S., Shinada, N. K., et al. (2020).
A structural entropy index to analyze conformations in intrinsically disordered proteins.
Journal of Structural Biology, 210, 107464.
Amman, B. R., Carroll, S. A., Reed, Z. D., Sealy, T. K., Balinandi, S., Swanepoel, R., et al.
(2012). Seasonal pulses of Marburg virus circulation in juvenile Rousettus aegyptiacus
bats coincide with periods of increased risk of human infection. PLoS Pathogens,
8(10), e1002877.
Ashour, H. M., Elkhatib, W. F., Rahman, M., & Elshabrawy, H. A. (2020). Insights into the
recent 2019 novel coronavirus (SARS-CoV-2) in light of past human coronavirus out-
breaks. Pathogens, 35(3), 235–241.
Baltimore, D. (1971). Expression of animal virus genomes. Bacteriological Reviews, 35(3),
235–241.
Barrı́a-Sandoval, C., Ferreira, G., Benz-Parra, K., & López-Flores, P. (2021). Prediction of
confirmed cases of and deaths caused by COVID-19 in Chile through time series tech-
niques: A comparative study. PLoS One, 16(4), e0245414.
Batschelet, E., Domingo, E., & Weissmann, C. (1976). The proportion of revertant and mutant
phage in a growing population, as a function of mutation and growth rate. Gene, 1(1),
27–32.
Becerra-Flores, M., & Cardozo, T. (2020). SARS-CoV-2 viral spike G614 mutation exhibits
higher case fatality rate. International Journal of Clinical Practice, 74, e13525.
Bernasconi, A., Mari, L., Casagrandi, R., & Ceri, S. (2021). Data-driven analysis of amino acid
change dynamics reveals SARS-CoV-2 variant emergence. Scientific Reports, 11, 21068.
70 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Berngruber, T. W., Froissart, R., Choisy, M., & Gandon, S. (2013). Evolution of virulence in
emerging epidemics. PLoS Pathogens, 9(3), e1003209.
Bjørnstad, O. N., & Viboud, C. (2016). Timing and periodicity of influenza epidemics. Pro-
ceedings of the National Academy of Sciences of the United States of America, 113(46),
12899–12901.
Bonhoeffer, S., Chappey, C., Parkin, N. T., Whitcomb, J. M., & Petropoulos, C. J. (2004). Ev-
idence for positive epistasis in HIV-1. Science, 306(5701), 1547–1550.
Borrmann, H., McKeating, J. A., & Zhuang, X. (2021). The circadian clock and viral infec-
tions. Journal of Biological Rhythms, 36, 9–22.
Brownlee, J. (1918). An investigation into the periodicity of measles epidemics in London
from 1703 to the present day by the method of the periodogram. Philosophical Transac-
tions of the Royal Society London B, 208, 225–250.
Burch, C. L., & Chao, L. (2004). Epistasis and its relationship to canalization in the RNA virus
φ6. Genetics, 167(2), 559–567.
Burra, P., Soto-Dı́az, K., Chalen, I., Gonzalez-Ricon, R. J., Istanto, D., & Caetano-Anolles, G.
(2021). Temperature and latitude correlate with SARS-CoV-2 epidemiological variables
but not with genomic change worldwide. Evolutionary Bioinformatics, 19,
1176934321989695.
Caetano-Anolles, G. (2021). The language of biomolecular communication. In G. Caetano-
Anolles (Ed.), Untangling molecular biodiversity (pp. 283–345). Singapore: World Scien-
tific Publishing.
Caetano-Anolles, D., & Caetano-Anolles, G. (2015). Computing the origin and evolution of
the ribosome from its structure—Uncovering processes of macromolecular accretion
benefitting synthetic biology. Computational and Structural Biotechnology Journal, 13,
427–447.
Caetano-Anolles, G., Mughal, F., Aziz, M. F., Koç, I., Caetano-Anolles, K., Caetano-Anolles,
D., et al. (2021). A ’double tale’ of module creation in evolving networks. In G. Caetano-
Anolles (Ed.), Untangling molecular biodiversity (pp. 91–168). Singapore: World Scien-
tific Publishing.
Cannell, J. J., Vieth, R., Umhau, J. C., Holick, M. F., Gran, W. B., Madronich, S., et al. (2006).
Epidemic influenza and vitamin D. Epidemiology and Infection, 134, 1129–1140.
Carlson, C. J., Gomez, A. C., Bansal, S., & Ryan, S. J. (2020). Misconceptions about weather
and seasonality must not misguide COVID-19 response. Nature Communications, 11(1),
1–4.
Chambliss, D. F., & Schutt, R. K. (2012). Making sense of the social world (4th ed.). London:
SAGE Publications.
Chin, A. W. H., Chu, J. T. S., Perera, M. R. A., Hui, K. P. Y., Yen, H.-L., Chan, M. C. W., et al.
(2020). Stability of SARS-CoV-2 in different environmental conditions. The Lancet Mi-
crobe, 1, e10.
Colominas, M. A., Schlotthauer, G., & Torres, M. E. (2014). Improved complete ensemble
EMD: A suitable tool for biomedical signal processing. Biomedical Signal Process and
Control, 14, 19–29.
Conlan, A. J. K., & Grenfell, B. T. (2007). Seasonality and the persistence and invasion of
measles. Proceedings of the Royal Society B: Biological Science, 274, 1133–1141.
Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. (2020).
The species severe acute respiratory syndrome-related coronavirus: Classifying 2019-
nCoV and naming it SARS-CoV-2. Nature Microbiology, 5, 536–544.
Cox, N. J., & Fukuda, K. (1998). Influenza. Infectious Disease Clinics of North America, 12,
27–38.
References 71
Demongeot, J., Flet-Berliac, Y., & Seligmann, H. (2020). Temperature decreases spread pa-
rameters of the new Covid-19 case dynamics. Biology (Basel), 9, 94.
Deyle, E. R., Maher, M. C., Hernandez, R. D., Basu, S., & Sugihara, G. (2016). Global envi-
ronmental drivers of influenza. Proceedings of the National Academy of Science of the
United States of America, 113, 13081–13086.
Dings, R. P. M., Miller, M. C., Griffin, R. J., & Mayo, K. H. (2018). Galectins as
molecular targets for therapeutic intervention. International Journal of Molecular
Sciences, 19, 905.
Domingo, E. (2002). Quasispecies theory in virology. Journal of Virology, 76(1), 463–465.
Domingo, E., Martin, V., Perales, C., Grande-Perez, A., Garcia-Arriaza, J., & Arias, A. (2006).
Viruses as quasispecies: Biological implications. Current Topics in Microbiology and
Immunology, 299, 51–82.
Domingo, E., Sheldon, J., & Perales, C. (2012). Viral quasispecies evolution. Microbiology
and Molecular Biology Reviews, 76, 159–216.
Domingo, E., Ruiz-Jarabo, C. M., Sierra, S., Arias, A., Pariente, N., Baranowski, E., et al.
(2002). Emergence and selection of RNA virus variants: Memory and extinction. Virus
Research, 82(1–2), 39–44.
Dopico, X. C., Evangelou, M., Ferreira, R. C., Guo, H., Pekalski, M. L., Smyth, D. J., et al.
(2015). Widespread seasonal gene expression reveals annual differences in human immu-
nity and physiology. Nature Communications, 6, 7000.
Dowell, J. (2001). Seasonal variation in host susceptibility and cycles of certain infectious dis-
eases. Emerging Infectious Diseases, 7(3), 369–374.
Drake, J. W., Charlesworth, B., Charlesworth, D., & Crow, J. F. (1998). Rates of spontaneous
mutation. Genetics, 148(4), 1667–1686.
Dudas, G., Carvalho, L. M., Rambaut, A., & Bedford, T. (2018). MERS-CoV spillover at the
camel-human interface. eLife, 7(e31257), 1–37.
Duncan, C. J., Duncan, S. R., & Scott, S. (1997). The dynamics of measles epidemics.
Theoretical Population Biology, 2, 155–163.
Dunker, A. K., Brown, C. J., Lawson, J. D., Iakoucheva, L. M., & Obradovic, Z. (2002).
Intrinsic disorder and protein function. Biochemistry, 41, 6573–6582.
Earn, D. J., Dushoff, J., & Levin, S. A. (2002). Ecology and evolution of the flu. Trends in
Ecology and Evolution, 17(7), 334–340.
Eckerle, L. D., Becker, M. M., Halpin, R. A., Li, K., Venter, E., Lu, X., et al. (2010). Infidelity
of SARS-CoV Nsp14-exonuclease mutant virus replication is revealed by complete
genome sequencing. PLoS Pathogens, 6(5), e1000896.
Elbe, S., & Buckland-Merrett, G. (2017). Data, disease and diplomacy: GISAID’s innovative
contribution to global health. Global Challenges, 1, 33–46.
Farah-Perez, A., Umaña-Villalobos, G., Picado-Barboza, J., & Anderson, E. P. (2020). An
analysis of river fragmentation by dams and river dewatering in Costa Rica. River Re-
search and Applications, 36, 1442–1448.
Ferguson, N. M., Cucunubá, Z. M., Dorigatti, I., Nedjati-Gilani, G. L., Donnelly, C. A., Basá-
ñez, M. G., et al. (2016). Countering the Zika epidemic in Latin America. Science,
353(6297), 353–354.
Fine, P. E. M., & Clarkson, J. A. (1982). Measles in Engand and Wales—I: An analysis of
factors underlying seasonal patterns. International Journal of Epidemiology, 11, 5–14.
Finke, L. L. (1792-1795). Versuch einer allgemeinen medicinisch -praktischen Geographie. 3
Vols. Leipzig: Weidmannschen Buchhandlung.
Finkel, Y., Mizrahi, O., Nachshon, A., Weingarten-Gabbay, S., Morgenstern, D., Yahalom-
Ronen, Y., et al. (2020). The coding capacity of SARS-CoV-2. Nature, 589, 125–130.
72 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Hethcote, H. W., Stech, H. W., & Van Den Driessche, P. (1981). Nonlinear oscillations in
epidemic models. SIAM Journal on Applied Mathematics, 40(1), 1–9.
Hirsch, A. (1883). Handbook of geographical and historical pathology. Acute infective
diseases: Vol. I. London: The New Sydenham Society.
Holm, L. (2020). DALI and the persistence of protein shape. Protein Science, 29, 128–140.
Hope-Simpson, R. E. (1981). The role of season in the epidemiology of influenza. Journal of
Hygiene (London), 86(1), 35–47.
Huang, Y., Yang, C., Xu, X.-F., Xu, W., & Liu, S.-W. (2020). Structural and functional prop-
erties of SARS-CoV-2 spike protein: Potential antivirus drug development for COVID-19.
Acta Pharmacologica Sinica, 41, 1141–1149.
Janeway, C., Travers, P., Walport, M., & Shlomchik, M. (2001). Immunobiology (5th ed.).
New York: Garland Science.
Johnson, T., & Barton, N. H. (2002). The effect of deleterious alleles on adaptation in asexual
populations. Genetics, 162(1), 395–411.
Jones, K. E., Patel, N. G., Levy, M. A., Storeygard, A., Balk, D., Gittleman, J. L., et al. (2008).
Global trends in emerging infectious diseases. Nature, 451, 990–993.
Jonsson, C. B., Moraes Figuereido, L. T., & Vapalahti, O. (2010). A global perspective on
Hantavirus ecology, epidemiology, and disease. Clinical Microbiology Reviews, 23(2),
412–441.
Karapiperis, C., Kouklis, P., Papastratos, S., Chasapi, A., Danchin, A., Angelis, L., et al.
(2021). A strong seasonality pattern for COVID-19 incidence rates modulated by UV
radiation levels. Viruses, 13, 574.
Kasahara, A. K., Singh, R. J., & Noymer, A. (2013). Vitamin D (25OHD) serum seasonality in
the United States. PloS One, 8, e65785.
Kassem, A. Z. E. (2020). Does temperature affect COVID-19 transmission? Frontiers in
Public Health, 8, 554964.
Khatchikian, D., Orlich, M., & Rott, R. (1989). Increased viral pathogenicity after insertion of
a 28S ribosomal RNA sequence into the haemagglutinin gene of an influenza virus.
Nature, 340(6229), 156–157.
Killerby, M. E., Biggs, H. M., Haynes, A., Dahl, R. M., Mustaquim, D., Gerber, S. I., et al.
(2018). Human coronavirus circulation in the United States 2014-2017. Journal of Clinical
Virology, 101, 52–56.
Kirchdoerfer, R. N., & Ward, A. B. (2019). Structure of the SARS-CoV NSP12 polymerase
bound to NSP7 and NSP8 co-factors. Nature Communications, 10, 2342.
Kissler, S. M., Tedijanto, C., Goldstein, E., Grad, Y. H., & Lipsitch, M. (2020). Projecting the
transmission dynamics of SARS-CoV-2 through the postpandemic period. Science,
368(6493), 860–868.
Kleypas, J. A., Castruccio, F. S., Curchitser, E. N., & McLeod, E. (2015). The impact of ENSO
on coral heat stress in the western equatorial Pacific. Global Change Biology, 21,
2525–2539.
Klingberg, E., Olerod, G., Konar, J., Petzold, M., & Hammarsten, O. (2015). Seasonal vari-
ations in serum 25-hydroxy vitamin D levels in a Swedish cohort. Endocrine, 49, 800–808.
Knight, G. (1971). The ecology of African sleeping sickness. Annals of the Association of
American Geographers, 61(1), 23–44.
Korber, B., Fischer, W. M., Gnanakaran, S., Yoon, H., Theiler, J., Abfalterer, W., et al. (2020).
Tracking changes in SARS-CoV-2 spike: Evidence that D614G increases infectivity of the
COVID-19 virus. Cell, 182, 812–827.
74 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Kronfeld-Schor, N., Stevenson, T. J., Nickbakhsh, S., Schernhammer, E. S., Dopico, X. C.,
Dayan, T., et al. (2021). Driversof infectious disease seasonality: Potential implications
for COVID-19. Journal of Biological Rythms, 36(1), 35–54.
Kvennefors, E. C., Leggat, W., Kerr, C. C., Ainsworth, T. D., Hoegh-Guldberg, O., &
Barnes, A. C. (2010). Analysis of evolutionarily conserved innate immune components
in coral links immunity and symbiosis. Developmental and Comparative Immunology,
34, 1219–1229.
Lauber, C., Goeman, J. J., Parquet, M. C., Thi Nga, P., Snijder, E. J., Morita, K., et al. (2013).
The footprint of genome architecture in the largest genome expansion in RNA viruses.
PLoS Pathogens, 9(7), e1003500.
Lauring, A. S., & Andino, R. (2010). Quasispecies theory and the behavior of RNA viruses.
PLoS Pathogens, 6(7), e1001005.
Li, R., Metcalf, J. E., Stenseth, N. C., & Bjørnstad, O. N. (2021). A general model for the de-
mographic signatures of the transition from pandemic emergence to endemicity. Science
Advances, 7, eabf9040.
Li, Y., Wang, X., & Nair, H. (2020). Global seasonality of human seasonal coronaviruses:
A clue for postpandemic circulating season of severe acute respiratory syndrome corona-
virus 2? Journal of Infectious Diseases, 222, 1090–1097.
Liu, X., Huang, J., Li, C., Zhao, Y., Wang, D., Huang, Z., et al. (2021). The role of seasonality
in the spread of COVID-19 pandemic. Environmental Research, 195, 110874.
Lowen, A. C., Mubareka, S., Steel, J., & Palese, P. (2007). Influenza virus transmission is
dependent on relative humidity and temperature. PLoS Pathogens, 3(10), 1470–1476.
Majumdar, P., & Niyogi, S. (2021). SARS-CoV-2 mutations: The biological trackway towards
viral fitness. Epidemiology and Infection, 149, e110.
Malim, M. H., & Emerman, M. (2001). HIV-1 sequence variation: Drift, shift, and attenuation.
Cell, 104(4), 469–472.
Manrubia, S. C., & Lázaro, E. (2006). Viral evolution. Physics of Life Reviews, 3(2), 65–92.
Mantilla-Beniers, N. B., Bjørnstad, O. N., Grenfell, B. T., & Rohani, P. (2010). Decreasing
stochasticity through enhanced seasonality in measles epidemics. Journal of the Royal
Society Interface, 7(46), 727–739.
Martin, D. P., Weaver, S., Tegally, H., San, E. J., Shank, S. D., Wilkinson, E., et al. (2021). The
emergence and ongoing convergent evolution of the N501Y lineages coincides with a
major global shift in the SARS-CoV-2 selective landscape. medRxiv. https://doi.org/
10.1101/2021.02.23.21252268.
Martinez, M. E. (2018). The calendar of epidemics: Seasonal cycles of infectious diseases.
PLoS Pathogens, 14(11), e1007327.
Matson, M. J., Yinda, C. K., Seifert, S. N., Bushmaker, T., Fischer, R. J., van Doremalen, N.,
et al. (2020). Effect of environmental conditions on SARS- CoV-2 stability in human nasal
mucus and sputum. Emergent Infectious Diseases, 26, 2276–2278.
Mecenas, P., Bastos, R. T. R. M., Vallinoto, A. C. R., & Normando, D. (2020). Effects of tem-
perature and humidity on the spread of COVID-19: A systematic review. PLoS One, 15(9),
e0238339.
Mercatelli, D., & Giorgi, F. M. (2020). Geographic and genomic distribution of SARS-CoV-2
mutations. Frontiers in Microbiology, 11, 1800.
Midgley, C. M., Haynes, A. K., Baumgardner, J. L., Chommanard, C., Demas, S. W.,
Prill, M. M., et al. (2017). Determining the seasonality of respiratory syncytial virus in
the United States: The impact of increased molecular testing. Journal of Infectious
Diseases, 216, 345–355.
References 75
Minhaz Ud-Dean, S. M. (2010). Structural explanation for the effect of humidity on persis-
tence of airborne virus: Seasonality of influenza. Journal of Theoretical Biology,
263(3), 822–829.
Minskaia, E., Hertzig, T., Gorbalenya, A. E., Campanacci, V., Cambillau, C., Canard, B., et al.
(2006). Discovery of an RNA virus 30 !50 exoribonuclease that is critically involved in
coronavirus RNA synthesis. Proceedings of the National Academy of Sciences of the
United States of America, 103, 5108–5113.
Montgomery, D. (2013). Design and analysis of experiments. Hoboken, NJ: John Wiley &
Sons, Inc.
Monto, A. S., DeJonge, P., Callear, A. P., Bazzi, L. A., Capriola, S., Malosh, R. E., et al.
(2020). Coronavirus occurrence and transmission over 8 years in the HIVE cohort of
households in Michigan. Journal of Infectious Diseases, 222, 9–16.
Morikawa, S., Kohdera, U., Hosaka, T., Ishii, K., Akagawa, S., Hiroi, S., et al. (2015). Seasonal
variations of respiratory viruses and etiology of human rhinovirus infection in children.
Journal of Clinical Virology, 73, 14–19.
Moriyama, M., Hugentobler, W. J., & Iwasaki, A. (2020). Seasonality of respiratory viral
infections. Annual Review of Virology, 7, 83–101.
Nasci, R. S., Savage, H. M., White, D. J., Niller, J. R., Cropp, B. C., Godsey, M. S., et al.
(2001). West Nile virus in overwintering Culex mosquitoes, New York City, 2000.
Emerging Infectious Diseases, 7, 742–744.
Nasir, A., Kim, K. M., & Caetano-Anolles, G. (2017). Long-term evolution of viruses:
A Janus-faced balance. BioEssays, 39, 1700026.
Nickbakhsh, S., Ho, A., Marques, D. F., McMenamin, J., Gunson, R. N., & Murcia, P. R.
(2020). Epidemiology of seasonal coronaviruses: Establishing the context for the
emergence of coronavirus disease 2019. Journal of Infectious Diseases, 222, 17–25.
Nora, T., Charpentier, C., Tenaillon, O., Hoede, C., Clavel, F., & Hance, A. J. (2007).
Contribution of recombination to the evolution of human immunodeficiency viruses
expressing resistance to antiretroviral treatment. Journal of Virology, 81(14), 7620–7628.
Paccaud, M. (1979). World trends in poliomyelitis morbidity and mortality, 1951-1975. World
Health Statistics Quarterly, 32, 198–224.
Pan, K., & Deem, M. W. (2011). Quantifying selection and diversity in viruses by entropy
methods, with application to the haemagglutinin of H3N2 influenza. Journal of the Royal
Society Interface, 8, 1644–1653.
Peng, G., Xu, L., Lin, Y. L., Chen, L., Pasquarella, J. R., Holmes, K. V., et al. (2012). Crystal
structure of bovine coronavirus spike protein lectin domain. Journal of Biological Chem-
istry, 287, 41931–41938.
Phan, T. (2020). Genetic diversity and evolution of SARS-CoV-2. Infection, Genetics and
Evolution, 81, 104260.
Poirier, C., Luo, W., Majumder, M. S., Liu, D., Mandl, K. D., Mooring, T. A., et al. (2020). The
role of environmental factors on transmission rates of the COVID-19 outbreak: An initial
assessment in two spatial scales. Scientific Reports, 10(1), 1–11.
Poole, L. (2020). Seasonal influences on the spread of SARS-CoV-2 (COVID19), causality,
and forecastabililty (3-15-2020). SSRN Electronic Journal. https://doi.org/10.2139/ssrn.
3554746.
Posada, D., Crandall, K. A., & Holmes, E. C. (2002). Recombination in evolutionary geno-
mics. Annual Review of Genetics, 36(1), 75–97.
Rahimi, N. (2021). C-type Lectin CD209L/L-SIGN and CD209/DC-SIGN: Cell adhesion
molecules turned to pathogen recognition receptors. Biology, 10, 1.
76 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Rahman, M. S., Islam, M. R., Rubayet Ul Alam, A. S. M., Islam, I., Hoque, M. N., Akter, S.,
et al. (2021). Evolutionary dynamics of SARS-CoV-2 nucleocapsid protein and its conse-
quences. Journal of Medical Virology, 93(4), 2177–2195.
Rath, S. L., & Kumar, K. (2020). Investigation of the effect of temperature on the structure of
SARS-CoV-2 Spike protein by molecular dynamics simulations. Frontiers in Molecular
Biosciences, 7, 583523.
Rhodes, T., Wargo, H., & Hu, W.-S. (2003). High rates of human immunodeficiency virus type
1 recombination: Near-random segregation of markers one kilobase apart in one round of
viral replication. Journal of Virology, 77(20), 11193–11200.
Ricci, C. A., Kamal, A. H. M., Chakrabarty, J. K., Fuess, L. E., Mann, W. T., Jinks, L. R., et al.
(2019). Proteomic investigation of a diseased Gorgonian coral indicates disruption of
essential cell function and investment in inflammatory and other immune processes. Inte-
grative and Comparative Biology, 59(4), 830–844.
Rodgers, J. L., & Nicewander, W. A. (1988). Thirteen ways to look at the correlation coeffi-
cient. The American Statistician, 43(1), 59–66.
Rohani, P., Earn, D. J., & Grenfell, B. T. (1999). Opposite patterns of synchrony in sympatric
disease metapopulations. Science, 286(5441), 968–971.
Sailani, M. R., Metwally, A. A., Zhou, W., Rose, S. M. S.-F., Ahadi, S., Contrepois, K., et al.
(2020). Deep longitudinal multiomics profiling reveals two biological seasonal patterns in
California. Nature Communications, 11, 4933.
Sajadi, M. M., Habibzadeh, P., Vintzileos, A., Shokouhi, S., Miralles-Wilhelm, F., &
Amoroso, A. (2020). Temperature, humidity, and latitude analysis to estimate potential
spread and seasonality of coronavirus disease 2019 (COVID-19). JAMA Network Open,
3, e2011834.
Sanjuán, R., & Domingo-Calap, P. (2016). Mechanisms of viral mutation. Cellular and Mo-
lecular Life Sciences, 73(23), 4433–4448.
Sanjuán, R., Moya, A., & Elena, S. F. (2004). The distribution of fitness effects caused by
single-nucleotide substitutions in an RNA virus. Proceedings of the National Academy
of Sciences of the United States of America, 101(22), 8396–8401.
Saputri, D. S., Li, S., van Eerden, F. J., Rozewicki, J., Xu, Z., Ismanto, H. S., et al. (2020).
Flexible, functional, and familiar: Characteristics of SARS-CoV-2 spike protein evolution.
Frontiers in Microbiology, 11, 2112.
Sato, S., St-Pierre, C., Bhaumik, P., & Nieminen, J. (2009). Galectins in innate immunity: Dual
functions of host soluble β-galactosidase-binding lectins as damage-associated molecular
patterns (DAMPs) and as receptors for pathogen-associated molecular patterns (PAMPs).
Immunological Reviews, 230, 172–187.
Schober, A. F., Rzhetsky, A., & Rust, M. J. (2021). Seasonal disease in the United States has
the hallmarks of an entrained circannual clock. medRxiv. https://doi.org/10.1101/2021.05.
26.21257655.
Seronello, S., Montanez, J., Presleigh, K., Barlow, M., Park, S. B., & Choi, J. (2011). Ethanol
and reactive species increase basal sequence heterogeneity of hepatitis C virus and produce
variants with reduced susceptibility to antivirals. PLoS One, 6(11), e27436.
Seyer, A., & Sanlidag, T. (2020). Solar ultraviolet radiation sensitivity of SARS-CoV-2. The
Lancet Microbe, 1(1), e8–e9.
Shaman, J., Pitzer, V. E., Viboud, C., Grenfell, B. T., & Lipsitch, M. (2010). Absolute humid-
ity and the seasonal onset of influenza in the continental United States. PLoS Biology, 8(2),
e1000316.
Sheftel, H., Shoval, O., Mayo, A., & Alon, U. (2013). The geometry of the Pareto front in
biological phenotype space. Ecology and Evolution, 3, 1471–1483.
References 77
Showers, W. M., Leach, S. M., Kechris, K., & Strong, M. (2021). Analysis of SARS-CoV-2
mutations over time reveals increasing prevalence of variants in the spike protein and
RNA-dependent RNA polymerase. bioRxiv. https://doi.org/10.1101/2021.03.05.433666.
Singh, J., Pandit, P., McArthur, A. G., Banerjee, A., & Mossman, K. (2021). Evolutionary tra-
jectory of SARS -CoV-2 and emerging variants. Virology Journal, 18, 166.
Smit, A. J., Fitchett, J. M., Engelbrecht, F. A., Scholes, R. J., Dzhivhuho, G., & Sweijd, N. A.
(2020). Winter is coming: A southern hemisphere perspective of the environmental drivers
of SARS-CoV-2 and the potential seasonality of COVID-19. International Journal of
Environmental Research and Public Health, 17(16), 5634.
Smith, E. C., & Denison, M. R. (2013). Coronaviruses as DNA wannabes: A new model for the
regulation of RNA virus replication fidelity. PLoS Pathogens, 9, e1003760.
Soh, W. T., Liu, Y., Nakayama, E. E., Ono, C., Torii, S., Nakagami, H., et al. (2021).
The N-terminal domain of spike glycoprotein mediates SARS-CoV-2 infection by
associating with L-SIGN and DC-SIGN. bioRxiv. https://doi.org/10.1101/2020.11.05.
369264.
Soper, H. E. (1929). The interpretation of periodicity in disease prevalence. Journal of the
Royal Statistical Society, 92(1), 34–73.
Soper, F. L. (1967). Dynamics of Aedes aegyptii distribution and density. Bulletin of the World
Health Organization, 36, 536–538.
Starr, T. N., Greaney, A. J., Hilton, S. K., Ellis, D., Crawford, K. H. D., Dingens, A. S., et al.
(2020). Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals
constraints on folding and ACE2 binding. Cell, 5, 1295–1310.
Sugihara, G., Grenfell, B., & May, R. M. (1990). Distinguishing error from chaos in ecological
time series. Philosophical Transactions of the Royal Society, London B, 330(1257),
235–251.
Sugihara, G., Nay, R., Ye, H., Hsieh, C.-H., Deyle, E., Fogarty, M., et al. (2012). Detecting
causality in complex ecosystems. Science, 338(6106), 496–500.
Tamerius, J., Nelson, M. I., Zhou, S. Z., Viboud, C., Miller, M. A., & Alonso, W. J. (2011).
Global influenza seasonality: Reconciling patterns across temperate and tropical regions.
Environmental Health Perspectives, 119, 439–445.
Tasakis, R. N., Samaras, G., Jamison, A., Lee, M., Paulus, A., Whitehouse, G., et al. (2021).
SARS-CoV-2 variant evolution in the United States: High accumulation of viral mutations
over time likely through serial founder events and mutation bursts. PLoS One, 16(7),
e0255169.
Thepaut, M., Luczkowiak, J., Vivès, C., Labiod, N., Bally, I., Lasala, F., et al. (2021).
DC/L-SIGN recognition of spike glycoprotein promotes SARS- CoV-2 trans-
infection and can be inhibited by a glycomimetic antagonist. PLoS Pathogens, 17(5),
e1009576.
Tomaszewski, T., DeVriers, R. S., Dong, M., Bhatia, G., Norsworthy, M. D., Zheng, X., et al.
(2020). New pathways of mutational change in SARS-CoV-2 proteomes involve regions
of intrinsic disorder important for virus replication and release. Evolutionary Bioinformat-
ics, 16, 1176934320965149.
Tosepu, R., Gunawan, J., Effendy, D. S., Ahmad, L. O. I. E., Lestari, H., Bahar, H., et al.
(2020). Correlation between weather and Covid-19 pandemic in Jakarta, Indonesia.
Science of the Total Environment, 725, 138436.
Troyano-Hernáez, P., Reinosa, R., & Holguı́n, Á. (2021). Evolution of SARS-CoV-2 enve-
lope, membrane, nucleocapsid, and spike structural proteins from the beginning of the pan-
demic to September 2020: A global and regional approach by epidemiological week.
Viruses, 13, 243.
78 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
van Doremalen, N., Bushmaker, T., Morris, D. H., Holbrook, M. G., Gamble, A.,
Williamson, B. N., et al. (2020). Aerosol and surface stability of SARS- CoV-2 as com-
pared with SARS-CoV-1. New England Journal of Medicine, 382, 1564–1567.
Vidal-Dupiol, J., Dheilly, N. M., Rondon, R., Grunau, C., Cosseau, C., Smith, K. M., et al.
(2014). Thermal stress triggers broad Pocillopora damicornis transcriptomic remodeling,
while Vibrio coralliilyticus infection induces a more targeted immuno-suppression
response. PLoS One, 9, e107672.
Vijaykrishna, D., Mujerki, R., & Smith, G. J. D. (2015). RNA virus reassortment: An evolu-
tionary mechanism for host jumps and immune evasion. PLoS Pathogens, 11(7),
e1004902.
Wang, C., Horby, P. W., Hayden, F. G., & Gao, G. F. (2020). A novel coronavirus outbreak of
global health concern. The Lancet, 395, 470–473.
Wang, M., Jiang, A., Gong, L., Lu, L., Guo, W., Li, C., et al. (2020). Temperature significant
change COVID-19 transmission in 429 cities. medRxiv. https://doi.org/10.1101/2020.02.
22.20025791.
Weber, T. P., & Stilianakis, N. I. (2008). Inactivation of influenza A viruses in the environment
and modes of transmission: A critical review. Journal of Infection, 57, 361–373.
Wessa, P. (2021). Free statistics software (v1.2.1), Office for Research Development and
Education. http://www.wessa.net.
Woo, J., Lee, E. Y., Lee, M., Kim, T., & Cho, Y.-E. (2019). An in vivo cell-based assay for
investigating the specific interaction between the SARS-CoV N-protein and its viral RNA
packaging sequence. Biochemistry and Biophysics Research Communications, 520,
499–506.
Wu, Y., Jing, W., Liu, J., Ma, Q., Yuan, J., Wang, Y., et al. (2020). Effects of temperature and
humidity on the daily new cases and new deaths of COVID-19 in 166 countries. Science of
the Total Environment, 729, 139051.
Wu, F., Zhao, S., Yu, B., Chen, Y. M., Wang, W., Song, Z. G., et al. (2020). A new coronavirus
associated with human respiratory disease in China. Nature, 579, 265–269.
Wu, Y., Zhou, Z., Wang, J., Luo, J., Wang, L., & Zhang, Y. (2019). Temperature regulates
the recognition activities of galectin to pathogen and symbiont in the sclerotinian coral
Pocillopora damicornis. Developmental and Comparative Immunology, 96, 103–110.
Wyse, C., O’Malley, G., Coogan, A., McConkey, S., & Smith, D. (2021). Seasonal and day-
time variation in multiple immune parameters in humans: Evidence from 329,261 partic-
ipants of the UK Biobank cohort. iScience, 24, 102255.
Xu, C., Wang, Y., Liu, C., Zhang, C., Han, W., Hong, X., et al. (2021). Conformational
dynamics of SARS-CoV-2 trimeric spike glycoprotein in complex with receptor ACE2
revealed by cryo-EM. Science Advances, 7(1), eabe5575.
Yafremava, L. S., Wielgos, M., Thomas, S., Nasir, A., Wang, M., Mittenthal, J. E., et al.
(2013). A general framework of persistence strategies for biological systems helps explain
domains of life. Frontiers in Genetics, 4, 16.
Yang, H.-Y., & Lee, J. K. W. (2021). The impact of temperature on the risk of COVID-19:
A multinational study. International Journal of Environmental Research and Public
Health, 18, 4052.
Ye, Q., West, A. M., Silletti, S., & Corbett, K. D. (2020). Architecture and self-assembly of the
SARS-CoV-2 nucleocapsid protein. Protein Science, 29(9), 1890–1901.
Zeger, S. L., Irizarry, R., & Peng, R. D. (2006). On time series analysis of public health and
biomedical data. Annual Reviews of Public Health, 27, 57–79.
Glossary 79
Zhou, L., Yang, H., Kuang, Y., Li, T., Xu, J., Li, S., et al. (2019). Temporal patterns of influ-
enza A subtypes and B lineages across age in a subtropical city, during pre-pandemic, pan-
demic, and postpandemic seasons. BMC Infectious Diseases, 19, 89.
Further reading
Callaway, E. (2020). The coronavirus is mutating-does it matter? Nature, 585(7824), 174–177.
Johannes, L., Jacob, R., & Leffler, H. (2019). Galectins at a glance. Journal of Cell Science,
131(9), jcs208884.
Li, F. (2016). Structure, function, and evolution of coronavirus spike proteins. Annual Review
of Virology, 3, 237–261.
Modenutti, C. P., Capurro, J. I. B., Di Lella, S., & Martı́, M. A. (2019). The structural biology
of galectin-ligand recognition: Current advances in modeling tools, protein engineering,
and inhibitor design. Frontiers in Chemistry, 7, 823.
Glossary
Amino acid variant A mutant protein with an amino acid replaced by another in a particular
site of its polypeptide sequence.
Baltimore classification of viruses A classification system of viruses that groups viruses
according to seven types of pathways used for mRNA synthesis: Group I: double-stranded
DNA viruses (dsDNA); Group II: single-stranded DNA viruses (ssDNA);Group III:
double-stranded RNA viruses (dsRNA); Group IV: positive sense single-stranded RNA
viruses (plus-ssRNA); Group V: negative sense single-stranded RNA viruses (minus-
ssRNA); Group VI: single-stranded reverse transcribing RNA viruses with a DNA inter-
mediate (ssRNA-RT); and Group VII: double-stranded reverse transcribing DNA viruses
with an RNA intermediate (dsRNA-RT).
Clade A group of taxa with a common evolutionary origin.
Coronavirus spike protein The spike (S) protein is the largest structural protein of corona-
viruses. The S-protein assemble into trimers to form spike structures that project outward
from the surface of the virion and give the typical ‘solar corona’ appearance of the virion
under negative stained transmission electron microscopy. The highly glycosylated
1200–1400 amino acid long protein is typically composed of two regions known as the
S1 and S2 subunits. The N-terminal S1 subunit interacts with receptor molecules on
the surface of cells and contains an N-terminal domain (NTD) and a C-terminal domain
(CTD). While both domains can interact with receptors, the CTD of the SARS-CoV-2
virus is known as the receptor-binding domain (RBD). The C-terminal S2 subunit embeds
the spike in the viral envelope and mediates viral entry by fusion of viral and host cell
membranes. It holds a ‘fusion’ region with hydrophobic fusion peptide (FP) and internal
fusion peptide (IFP) sequences, a ‘fusion core’ region composed of two heptad repeat (HR)
sequences that undergo major fusion-triggered conformational changes, and a C-terminal
transmembrane (TM) domain that acts as anchor.
Economy The budget of a system in terms of matter-energy costs to maintain its functioning
mechanisms.
Epidemic calendar A calendar of seasonality of viral and bacterial diseases.
80 CHAPTER 2 Seasonal behaviour of COVID-19 and its galectin-like culprit
Flexibility The structural and functional mechanisms responding to ‘internal’ and ‘external’
changes imposed on the system; these mechanisms require processing of information and
delimit the potential of the system to adapt to environmental change.
Galectins A class of proteins known to bind specifically to β-galactoside sugars with broad
specificity and harbour a disulphide bond-dependency for carbohydrate binding and
stability, making them S-type lectins.
Human coronaviruses (HCoVs) A group of coronaviruses that can cause infections of the
respiratory system and mild-to-severe illnesses, including common cold, pneumonia, se-
vere acute respiratory syndrome, kidney failure and even death.
Immune escape The ability of a virus to evade innate or adaptive immunity mechanisms.
Morphospace Spaces describing worlds of phenotypes, including observable molecular, cel-
lular, and organismal characteristics resulting from the interaction of genotypes with the
environment.
Persistence Maintenance of identity through time, including the continuance of a feature or
lineage of an organism or virus.
Phylogenetic tree A specific hypothesis of history (a phylogeny) in the form of a network
without reticulations.
Phylogeny A hypothesis of history and genealogical relationship among a group of entities
(taxa) in the form of a tree or network with specific connotations of ancestry and an implied
time axis.
Protein Intrinsic Disorder (PID) The lack or fixed or ordered three-dimensional molecular
structure that endows a molecule with flexibility, typically in the absence of macromolec-
ular interacting partners. PID can be a property of the entire protein in intrinsically disor-
dered proteins (IDPs) or of particular regions within protein domains or in linker or
terminal sequences. PID can be predicted with high confidence at per-residue level directly
from amino acid sequence with a number of algorithmic implementations, such as the
IUPred energy-based prediction method.
Robustness The mechanisms that use information to passively maintain structure and func-
tion despite external influence; these mechanisms protect the system from malfunction by
resisting damage and change.
Root-mean-square deviation (RMSD) When referring to atomic positions, RMSD measures
the average distance (in Å) between the atoms of superimposed proteins. Typically,
RMSDs of the Cα atomic coordinates of the protein backbone are used to measure simi-
larities of three-dimensional structures after optimal rigid body superposition. RMSD is
used as quantitatively measure of structural similarity between one or more proteins, with
lower values implying better structural match.
SARS-CoVs Strains of coronaviruses that cause severe acute respiratory syndrome (SARS), a
respiratory illness that materialized in two major outbreaks, the 2002–2004 SARS-CoV-1
outbreak and the ongoing 2019 SARS-CoV-2 pandemic outbreak.
Seasonal driver Causative agent of the seasonal behaviour of infectious diseases.
Seasonal forcing Seasonal variations in the onset and spread of infectious diseases.
Spike protein A round, flattened or button-shaped glycoprotein (previously known as a peplo-
mer protein; from Greek, peplos ‘robe’, meros ‘part’) that projects outward from the lipid
bilayer of the surface of an enveloped virus. Spike proteins play important roles, including
attachment of the virion to receptor sites on cell surfaces, mediating release of the nucle-
ocapsid with its genetic material, and harbouring hemagglutinating or enzymatic activities.
Glossary 81
Variant of concern (VOC) A variant of a virus exhibiting a set of mutations (amino acid var-
iants) that increase virus transmissibility, morbidity, mortality, risk, immune escape, diag-
nostic test evasion, or other criteria of significance.
Viral quasispecies A dynamical virus collective that is structured at a population level by
exhibiting a large number of variants that arise continually via mutation and are subjected
to natural selection.
Virion The infective submicroscopic particle form of a virus, complete with a capsid protein
coat structure encasing an infectious nucleic acid.
Virocell A cell with physiologies transformed by viral infection geared towards production of
virions rather than cellular multiplication.
Z-score In bioinformatics, the Z-score is simply the comparison of an actual alignment score
with the scores obtained on a set of random sequences by a Monte-Carlo process. In the
case of DALI, the Z-score is an optimized similarity score defined as the sum of equivalent
Cα-Cα atomic distances among two proteins. The maximum of the total score defines a
common structural core where every atom in the alignment makes a net positive contri-
bution. Scores are rescaled according to length because large structural alignments get
higher scores than smaller alignments. Significant similarities have Z-scores > 2, which
usually correspond to similar folds.
This page intentionally left blank
CHAPTER
Current molecular
diagnostics assays for
SARS-CoV-2 and
emerging variants
3
Jonathan M. Banksa, Kristelle Capistranoa, Pari Thakkara, Hemangi Ranadeb,
Vaidik Sonia, Manali Dattab, and Afsar R. Naqvia,*
a
Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL,
United States
b
Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, Rajasthan, India
*Corresponding author: e-mail address: [email protected]
Abbreviations
SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
WHO World Health Organization
VOC variant of concern
B.1.1.7 SARS-CoV-2 alpha variant
B.1.351 SARS-CoV-2 beta variant
P.1 SARS-CoV-2 gamma variant
B.1.617.2 SARS-CoV-2 delta variant
E protein envelope protein
S protein spike protein
M protein membrane protein
N protein nucleocapsid protein
RBD receptor-binding domain
ACE2 angiotensin-converting enzyme 2
NTD N-terminal domain
RT-PCR real-time reverse transcription polymerase chain reaction
cDNA complementary DNA
USFDA United States Food & Drug Administration
EUA emergency use authorization
CDC United States Centers for Disease Control and Prevention
LOD limit of detection
SGTF spike-gene target failure
LFIA lateral flow immunoassay
1 Introduction
In December 2019, the Severe Acute Respiratory Syndrome Coronavirus 2, better
known as SARS-CoV-2, underwent zoonotic transmission to infect a human and
cause a viral outbreak (Peñarrubia et al., 2020). On March 11, 2020, the World
Health Organization (WHO) declared COVID-19 a pandemic (van Dorp et al.,
2020). SARS-CoV-2 is genetically similar to a few other beta coronaviruses that in-
fect animals, including bats (RaTG13) and pangolins (Salian et al., 2021). Believed
to have originated due to mutation, SARS-CoV-2 spread from human to human and
continued mutating as it rapidly divided in each host (van Dorp et al., 2020). With the
world unable to contain SARS-CoV-2 or the surfacing mutants, the COVID-19 out-
break escalated to an epidemic and ultimately a pandemic, with high infection and
death rates globally (Salian et al., 2021).
On November 12, 2021, the World Health Organization COVID-19 Dashboard
reported 251,788,329 confirmed cases of COVID-19 as well as 5,077,907 deaths
from the disease (WHO COVID-19 Dashboard, 2020). The virus spreads through
air droplets, resulting in unique transmission patterns (Datta, Singh, & Naqvi,
2021). Countries worldwide experienced wave after wave of new cases, often fol-
lowed by a chaotic spike in deaths (Dyer, 2021). To manage and prevent new
COVID-19 waves, researchers must continue to study SARS-CoV-2 and develop
COVID-19 diagnostic methods to contain the viral spread (Vandenberg, Martiny,
Rochas, van Belkum, & Kozlakidis, 2020).
SARS-CoV-2 possesses a spherical shape of approximately 150 nm, and its
genome is 30 kb long single-stranded RNA (Datta et al., 2021). The viral genome
was sequenced in its entirety and shared with the NCBI Genbank on January 5, 2020.
2 SARS-CoV-2 variants of concern 85
has shown to confer one or more of the following viral attributes: increased transmis-
sibility, increased disease severity, decreased neutralizing antibody response acquired
through natural infection or vaccination, decreased treatment/vaccine efficacy, and
diagnostic detection failures.
Although mutations have accumulated throughout SARS-CoV-2 genes encoding
the ORF1ab, ORF3, ORF8, and N (nucleocapsid) proteins, mutations in the S (spike)-
protein are particularly significant. The S-protein is a type I transmembrane protein
that initially exists as an inactive precursor. During the early stages of infection,
SARS-CoV-2 uses its S-protein’s receptor-binding domain (RBD) to engage the
angiotensin-converting enzyme 2 (ACE2) host receptor, which is primarily expressed
in lung and intestinal cells. Following the RBD-ACE2 binding, proteolysis occurs at
the S1/S2 junction by furin or other proteases (i.e. TMPRSS2 or cathepsin proteases),
exposing a second cleavage site within the S2, subsequently liberating the S2 fusion
peptide to initiate the viral-host membrane fusion and leading to S1 shedding (Peacock
et al., 2021). Specific mutations such as D614G result in less S1 shedding and
increased incorporation of the S protein into the virion, thereby enhancing viral
infectivity.
Because the S-protein plays a crucial role in viral invasion and host cell attach-
ment, it is a major target for antibody therapy, specifically neutralizing antibodies
that target SARS-CoV-2. Within the S protein, the RBD and the NTD (N-terminal
domain) are primary targets. NTD-targeting and RBD-targeting antibodies form
complexes with the NTD and RBD, respectively, preventing viral entry and host
cell-virus membrane fusion. However, some variants such as B.1.351 (Beta) and
88 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
P.1 (Gamma) have developed mutations that prevent or weaken the binding of
particular antibodies to the S-protein (Yang & Du, 2021). This section highlights
changes in the viral genome, spread, and detection of each VOC.
higher than those infected with the original strain) further heightens the transmissi-
bility of B.1.617.2 (Delta) and could play a part in breakthrough infections (Li et al.,
2021; Reardon, 2021). Finally, one study found that B.1.617.2 (Delta) remains
infectious for longer (B.1.617.2 (Delta): 18 days; Wuhan-Hu-1: 13 days) (Ong
et al., 2021). Altogether, these data indicate that B.1.617.2 (Delta) replicates faster
and at higher rates compared to other mutants.
There are 12 mutations in the B.1.617.2 (Delta) genome, 10 of which are in the
S-protein: T19R, G142D (in some strains), 156del, 157del, R158G, L452R, T478K,
D614G, P681R, and D950N (Fig. 1). L452R and T478K both confer a significant
increase in immune evasion and infectivity to the B.1.617.2 (Delta) variant, and
they interfere with the host antibody response (monoclonal antibodies and sera from
convalescent and vaccinated individuals) by weakening the binding between anti-
bodies and the S-protein (Di Giacomo, Mercatelli, Rakhimov, & Giorgi, 2021;
Motozono et al., 2021; Planas et al., 2021). P681R is located in the S-protein
S1/S2 (furin) cleavage site, and it is another mutation that confers better transmis-
sibility (Cherian et al., 2021).
The VOCs share key mutations, mainly in the RBD and NBD-containing S1 sub-
unit. Consequently, a significant portion of these major spike mutations increases the
ACE2 binding affinity, altering viral pathogenicity and virulence. Not to mention,
specific mutations appear to have compensatory effects when combined. One exam-
ple is the K417N: N501Y combination seen in B.1.351 (Beta). K417N reportedly
reduces ACE2 binding affinity by disrupting the formation of two salt bridges in
the RBD-ACE2 complex. Despite K417N, B.1.351 (Beta) is still estimated to be
2.5 times more transmissible than Wuhan-Hu-1 due to the N501Y mutation
(Harvey et al., 2021).
The effect of these mutations on vaccine efficacy sensitivity is of great concern.
While SARS-CoV-2 vaccines mRNA-1273 (Moderna) and BNT162b2/Comirnaty
(Pfizer/BioNTech) reportedly elicit a robust antibody response against B.1.1.7
(Alpha) and P.1 (Gamma), the neutralization effect is significantly weaker against
B.1.351 (Beta). Data regarding the effect of B.1.617.2 (Delta) on the vaccines is
limited. However, early reports suggest that while protection against infection is
dampened, mRNA-1273 and BNT162b2 still vigorously protect against hospitaliza-
tion and death (Khateeb, Li, & Zhang, 2021).
3 COVID-19 diagnostics
A prevalent concern is the effect of mutations on diagnostic sensitivity. With the
background provided regarding the SARS-CoV-2 virus and its variants, the diagnos-
tic field is rapidly evolving. In this text, we discuss the various widely used diag-
nostic strategies that researchers developed to test for the virus and its effects on
the body (as manifested through antibody response). The sections below describe
four SARS-CoV-2 diagnostics approaches: RT-PCR, Serology, CRISPR and Bio-
sensor/electric/smart sensors (Fig. 2). Additionally, we discuss how diagnostic tech-
niques have been designed and redesigned for the detection of SARS-CoV-2.
92 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
FIG. 2
COVID-19 Diagnostic Techniques and their Targets (BioRender.com, 2021).
viral assays and diagnose pathogens (Deyde, Sampath, & Gubareva, 2011; Wolk,
Kaleta, & Wysocki, 2012). As with other pathogen diagnostics, RT-PCR tests play
a significant role in providing public health experts and government officials with
valuable information regarding viral presence and spread (Farasani, 2021).
At its core, RT-PCR involves the following processes: RNA is reverse tran-
scribed into complementary DNA (cDNA), then the cDNA is replicated exponen-
tially with each experimental cycle, and finally the expression levels of the cDNA
are observed in real time by fluorescent indicators as a proxy, and the expression
levels are used to extrapolate information regarding the genetic expression and
activity within a sample (Nolan et al., 2006). These samples can come from a variety
of sources, as previously mentioned, and PCR tests can probe for the presence of
several known pathogens (Fig. 3).
FIG. 3
RT-PCR Diagnostic Process. Schematic showing the description of obtaining a sample,
isolating RNA and RT-qPCR using instrument to detect viral genome (BioRender.com, 2021).
94 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
sensitivity, all of which make it a popular choice for diagnosing SARS-CoV-2 world-
wide (Fomsgaard & Rosenstierne, 2020). The accuracy of commercial RT-PCR tests
for COVID-19 were experimentally validated, and the tests were found to meet in-
ternational standards for accuracy (Wu, Xu, Zhu, & Xia, 2020). In addition to detect-
ing viral presence in samples, RT-PCR tests have yielded valuable information about
viral load and the infectivity of different COVID-19 variants (Korber et al., 2020).
Though they are not considered true rapid tests, in comparison to other methods used
for measuring RNA expression, PCR reactions are faster and safer, requiring fewer
dangerous materials for the tests (Farasani, 2021; Green & Sambrook, 2018).
There are several RT-PCR assays that governing bodies have approved for use in
testing for COVID-19. To rapidly respond to the developing COVID-19 pandemic,
the United States Food & Drug Administration (USFDA) issued Emergency Use
Authorizations (EUA) to quickly authorize the use of RT-PCR diagnostics. Accord-
ing to the USFDA, 210 RT-PCR diagnostic assays have been granted such approval
at the time of writing (https://www.fda.gov/medical-devices/coronavirus-disease-
2019-covid-19-emergency-use-authorizations-medical-devices/in-vitro-diagnos
tics-euas-molecular-diagnostic-tests-sars-cov-2). Two such tests were developed by
the United States Centers for Disease Control and Prevention (CDC), and the tests
fall into two separate categories of RT-PCR diagnostic assays: simplex and multiplex
assays (CDC, 2021). As detailed in Table 2, both tests target the nucleocapsid
(N) protein-coding gene in SARS-CoV-2. However, the multiplex assay also tests
for genes in the Influenza A and B viruses, allowing it to test for co-infection of
the viruses that manifest themselves similarly to COVID-19 (Shu et al., 2021).
Table 2 depicts their sensitivities as both >95%. The TaqPath COVID-19 Combo
Kit by Thermo Fisher tests for open reading frames 1a and 1b, the spike protein-
coding region, and the N protein-coding region of the SARS-CoV-2 genome
(www.thermofisher.com/us/en/home/clinical/clinical-genomics/pathogen-detection-
solutions/covid-19-sars-cov-2/multiplex.html; https://www.fda.gov/media/136112/
download). With a 100% detection rate at the limit of detection (LOD), this highly
sensitive test can detect various viral mutants because of its multi-site screening ca-
pabilities. Similarly, Labcorp’s COVID-19 RT-PCR Test is able to screen for mul-
tiple parts of the viral genome, resulting in a high sensitivity of >95% at the LOD
(https://www.fda.gov/media/136151/download). However, the invasive nature of
the nasal or bronchial swab required for these assays can cause subject discomfort.
Lastly, the most recent assay to be granted EUA approval at the time of writing
came out of the Yale School of Public Health, the SalivaDirect dualplex RT-PCR
assay. This assay yields comparable sensitivity of 94% agreement when compared
to the TaqPath assay, and its less invasive salivary sample requirement provides a
significant advantage without completely sacrificing sensitivity (https://ysph.yale.
edu/salivadirect/publications/; Vogels et al., 2021). These assays are described in
more detail in Table 2. Alternative RT-PCR techniques with fewer required reagents
were developed in response to a shortage in key RT-PCR reagents, and articles by
Fomsgaard and Rosenstierne (2020) and Smyrlaki et al. (2020) provide a more com-
prehensive description of these alternate approaches.
Table 2 RT-PCR diagnostic assays and their gene targets, sensitivities, limits of detection, and other key details.
Approval Sensitivity and limit of
Type of test, company, source and date Targets detection (LOD) Pros and cons
Influenza SARS-CoV-2 Multiplex Assay USFDA SARS-CoV-2 N gene, greater than 95% detection rate Pros: Tests
(Multiplex), CDC (CDC, 2021; https://www.fda. EUA, Influenza A Matrix (M1) gene, at LOD multiple viruses,
gov/medical-devices/coronavirus-disease-2019- 07/02/ Influenza B nonstructural 2 LOD ¼ Influenza A 102.0TCID50. high sensitivity
covid-19-emergency-use-authorizations- 2020 (NS2) gene Influenza B 10–0.1 EID50, and Cons: Limited to
medical-devices/in-vitro-diagnostics-euas- SARS-CoV-2 10–2.0 TCID50 genetic
molecular-diagnostic-tests-sars-cov-2; Shu et al., screening, time
2021) consuming
2019-nCoV RT-PCR Diagnostic Panel – QIAGEN USFDA SARS-CoV-2 N gene greater than 95% detection with Pros: High
EZ1 Advanced XL (Singleplex), CDC (CDC, 2021; EUA, LOD 100.5 RNA copies/μL sensitivity and
https://www.fda.gov/medical-devices/ 02/04/ accuracy
coronavirus-disease-2019- 2020 Cons: Tests only
covid-19-emergency-use-authorizations- one gene
medical-devices/in-vitro-diagnostics-euas-
molecular-diagnostic-tests-sars-cov-2)
TaqPath COVID-19 Combo Kit (Singleplex), USFDA SARS-CoV-2 ORF 1a, ORF 100% detection rate at LOD Pros: Tests
Thermo Fisher Scientific, (https://www.fda.gov/ EUA, 1b, S, and N genes LOD ¼ 10 Genomic Copy multiple genes,
medical-devices/coronavirus-disease-2019- 03/13/ Equivalents (GCE) per reaction, or differentiates
covid-19-emergency-use-authorizations- 2020 50 GCE/mL in 17.5 and 14.0 μL variants
medical-devices/in-vitro-diagnostics-euas- samples Cons: Tests for
molecular-diagnostic-tests-sars-cov-2; www. one virus, time
thermofisher.com/us/en/home/clinical/clinical- consuming
genomics/pathogen-detection-solutions/covid-
19-sars-cov-2/multiplex.html; https://www.fda.
gov/media/136112/download)
COVID-19 RT-PCR Test (Singleplex or Multiplex), USFDA SARS-CoV-2 nucleocapsid greater than 95% detection rate Pros: Targets
Laboratory Corporation of America (Labcorp) EUA, (N) gene at LOD multiple genes,
(https://www.fda.gov/medical-devices/ 03/16/ LOD ¼ 6.25 genome copies per versatile
coronavirus-disease-2019- 2020 microliter (cp/μL) for singleplex Cons: Only tests
covid-19-emergency-use-authorizations- and multiplex format one gene,
medical-devices/in-vitro-diagnostics-euas- invasive swab
molecular-diagnostic-tests-sars-cov-2; https://
www.fda.gov/media/136151/download)
SalivaDirect (Dualplex), Yale School of Public USFDA SARS-CoV-2 nucleocapsid 94% positive agreement when Pros: Efficient,
Health (https://www.fda.gov/medical-devices/ EUA, (N) gene compared to TaqPath COVID-19 noninvasive
coronavirus-disease-2019- 08/27/ combo kit, no LOD specified swab sample
covid-19-emergency-use-authorizations- 2021 Cons: Slightly
medical-devices/in-vitro-diagnostics-euas- lower sensitivity
molecular-diagnostic-tests-sars-cov-2; https://
ysph.yale.edu/salivadirect/publications/Vogels
et al., 2021)
96 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
either due to difficulty or no longer being present at the target site, as well as for
epidemiological and post-vaccination monitoring studies (Zhang et al., 2020). Sero-
logical tests can also aid with autoimmune disorder diagnosis, in the case of the body
mistaking its own healthy tissues for foreign invaders thus resulting in unnecessary
antibody production.
There are several types of serological tests, including: rapid diagnostic tests,
enzyme-linked immunosorbent assays, chemiluminescent immunoassays, electro-
chemiluminescence immunoassays, lateral flow chromatographic immunoassays
and neutralization assays (Datta et al., 2021). Rapid serology diagnostic tests (e.g.
lateral flow immunoassay [LFIA]) are convenient to implement at point-of-care,
rapid and inexpensive, thus allowing for commercial kit production (Yamamoto
et al., 2021). However, there is still a considerable amount of uncertainty with accu-
racy of rapid serologic tests.
Enzyme-linked immunosorbent assays (ELISA) rely on antigen binding to anti-
bodies in a laboratory (https://www.healthline.com/health/elisa). Chemiluminescent
immunoassays (CLIA) are also laboratory-based and rely on enzyme labelled
antibodies and antigens (https://www.lornelabs.com/news-events/blog/what-is-
chemiluminescent-immunoassay). One advantage of this assay technique is that
its sensitivity levels are very high. Electrochemiluminescence immunoassays
(ECLIA) rely on quantifying antigen or antibody, based on the change in electro-
chemiluminescence (ECL) signal, both before and after immunoreaction. ECLIA
has increased sensitivity and can be more selective than other methods (Wu & Ju,
2012). Lateral flow chromatographic immunoassays rely on the collected specimen’s
IgM or IgG antibodies binding to labelled SARS-CoV-2 antigens to form a complex,
which will later be accounted for (https://www.fda.gov/media/144071/download).
Neutralization assays measure the levels of neutralizing antibodies from either recent
or prior infection, which help inform the reasoning behind existing antibodies (https://
www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-
authorizes-first-test-detects-neutralizing-antibodies-recent-or). Due to lower sensi-
tivity of serological tests compared to molecular methods, serology tests are primarily
used for retrospective diagnosis (Datta et al., 2021).
SARS-CoV-2 has four structural surface proteins: E, S, M and N proteins (enve-
lope, spike, membrane, and nucleocapsid, respectively) (Datta et al., 2021). Serology
tests against SARS-CoV-2 have been created by several companies and have been
given emergency-use approval (EUA) via the U.S. Food and Drug Administration
(FDA) (Zhang et al., 2020). Other countries that have also similarly approved sero-
logical tests include Germany, Singapore, China, Japan and Spain. Most serological
tests have been developed to detect the presence of IgM/IgG antibodies against N,
S and Receptor-Binding Domain (RBD) proteins in human plasma and serum
(Ishikawa et al., 2009). Therefore, each serological test may be used for a single
or a few variants, until a new variant emerges with mutations in the protein targeted
by the serology test (https://publichealth.jhu.edu/2021/variants-vaccines-and-what-
they-mean-for-covid-19-testing). As a result, serology diagnostics should not be
used for vaccine efficacy testing, due to different vaccine targets. However, with
3 COVID-19 diagnostics 99
further testing, serological diagnostic tests for COVID-19 may be used to inform a
level of immunity that could perhaps decrease the severity of re-infection and the
time during which the infection remains.
There are several serology diagnostic assays that are currently being studied for
use against COVID-19 and its several variants (Table 3). FDA authorizes each for
use under the Emergency Use Authorization (EUA). The Elecsys AntiSARS-
CoV-2 S test uses an electrochemiluminescence quantitative immunoassay, and its
overall sensitivity is 99.5%, post 14 days of PCR confirmation (https://www.
centerforhealthsecurity.org/covid-19TestingToolkit/testing-basics/types-of-COVID-19-
tests/serology-tests.html). SARS-CoV-2 antibodies against the spike RBD (Receptor
Binding Domain) of the spike (S) protein (https://diagnostics.roche.com/us/en/
products/params/elecsys-anti-sars-cov-2.html; https://publichealth.jhu.edu/2021/
variants-vaccines-and-what-they-mean-for-covid-19-testing).
The Innovita 219-nCoV Ab Test (Colloidal Gold) employs a rapid lateral flow
qualitative chromatographic immunoassay, and its sensitivity is about 100%, with
its confidence interval of (88.7%, 100%) (https://www.centerforhealthsecurity.
org/covid-19TestingToolkit/testing-basics/types-of-COVID-19-tests/serology-tests.
html). This test uses antibodies against both the S1 and nucleocapsid (N) proteins of
SARS-CoV-2 (https://www.fda.gov/media/144071/download), as the antigens from
these regions are colloidal gold-labelled. Although mutations are possible in both
regions, testing for antibodies that match two different proteins results in a higher
efficacy.
The Dimension EXL SARS-CoV-2 Total antibody assay (CV2T) is an
example of a quantitative chemiluminescent immunoassay, and its sensitivity
after early SARS-CoV-2 infection in the host is unknown (https://www.cen
terforhealthsecurity.org/covid-19TestingToolkit/testing-basics/types-of-COVID-
19-tests/serology-tests.html). The target antigen of this assay is the S1 protein, which
has been associated with several possible mutations that could potentially lead to
several COVID-19 variants (Fig. 1) (https://www.fda.gov/media/138757/down
load). Access SARS-CoV-2 IgM test also relies on a chemiluminescent immunoas-
say and its sensitivity after early infection is unknown and requires further study.
This test can also yield false negative results if the quantity of antibodies against
the SARS-CoV-2 virus is too low or if the virus has acquired one or more amino acid
mutations in the RBD of the viral S1 protein recognized by the antibodies employed
in this test. These challenges result in decreased assay efficacy (https://www.
beckmancoulter.com/products/immunoassay/access-sars-cov-2-igm-antibody-test).
The OmniPATH COVID-19 Total Antibody Test is a quantitative enzyme-linked
immunosorbent assay (ELISA), and its overall sensitivity is 100%, post 15 days since
symptom onset (https://www.centerforhealthsecurity.org/covid-19TestingToolkit/
testing-basics/types-of-COVID-19-tests/serology-tests.html). Similar to other serol-
ogy tests, this test can also result in false negative results. Since this assay employs
SARS-CoV-2 antibodies against the RBD of the S1 protein, its accuracy is also
dependent on the possible mutations of this region in COVID-19 disease variants
(https://www.thermofisher.com/covid-19-antibody-testing/us/en/solutions/Omni
PATH-COVID19-Total-Antibody-ELISA-Test.html). The Platelia SARS-CoV-2
Table 3 Serology diagnostic assays and their sensitivity ranges, their target proteins, and several other key information.
Quantitative
Kits available, or
Type of test, source Company, Approval Targets Sensitivity qualitative Pros and cons
Total Ab-ECLIA (Datta et al., 2021; https:// Elecsys AntiSARS-CoV- SARS-CoV-2 Ag 14 days post PCR confirmation (95% Cl): Quantitative Pros: Takes very
diagnostics.roche.com/us/en/products/ 2 S, Roche Diagnostics, against spike 99.5% (97.0–100%) little time (18 min),
params/elecsys-anti-sars-cov-2.html; USFDA EUA (S) protein RBD epidemiological
https://diagnostics.roche.com/global/en/ application
news-listing/2020/roche-develops-new- Cons: Only used
serology-test-to-detect-covid- in vitro to date,
19-antibodies.html) prone to false
positives due to
pre-existing
antibodies
Lateral flow IgM/G (Datta et al., 2021; https:// Innovita 219-nCoV Ab Test SARS-CoV-2 Ag 100% (30/30) with confidence interval of Qualitative Pros: Aids with
www.fda.gov/media/144071/download) (Colloidal Gold), Innovita against S1 and (88.7%, 100%). Possible lower sensitivity to variant detection
(Tangshan) Biological nucleocapsid IgG antibodies in symptomatic individuals less Cons: Limited to
Technology Co., Ltd., (N) proteins than 15 days since symptom onset. Sensitivity IgM, IgG antibodies,
USFDA EUA of this test after early infection is unknown prescription only
Total antibody, CLIA (Datta et al., 2021; Dimension EXL SARS- SARS-CoV-2 Ag Sensitivity of this test after early infection is Quantitative Pros: Low
https://www.fda.gov/media/138757/ CoV-2 Total antibody against S1 unknown probability of false
download) assay (CV2T), Siemens protein RBD positives, detects
Healthcare Diagnostics total antibody levels
Inc., USFDA EUA Cons: Prescription
use only, complex
assay
IgM-CLIA (https://www.fda.gov/media/ Access SARS-CoV-2 IgM, SARS-CoV-2 Ag Sensitivity of this test after early infection is Quantitative Pros: Low
142911/download; https://www. Beckman Coulter, Inc., against S1 unknown maintenance, easy
beckmancoulter.com/products/ USFDA EUA protein RBD workflow integration
immunoassay/access-sars-cov-2- Cons: Limited to
igm-antibody-test; Datta et al., 2021) IgM antibodies,
prone to false
positives
Total Ab-ELISA (Datta et al., 2021; https:// OmniPATH COVID-19 SARS-CoV-2 Ag 15 days post symptom onset (95% Cl): Quantitative Pros: Detects IgM,
www.thermofisher.com/covid-19- Total Antibody ELISA Test, against S1 100% (89.2, 100.0). Sensitivity of this test after IgA, IgG bound
antibody-testing/us/en/solutions/ Thermo Fisher Scientific, protein RBD early infection is unknown antibodies
OmniPATH-COVID19-Total-Antibody- USFDA EUA Cons: Only used
ELISA-Test.html; https://www.fda.gov/ in vitro to date,
media/142700/download) prone to false
positives
Total Ab-ELISA (Datta et al., 2021; https:// Platelia SARS-CoV-2 Total SARS-CoV-2 Ag Overall: 49/50 (98%); 95% CI: 89.51–99.65% Quantitative Pros: One-step
www.fda.gov/media/137493/download; Ab assay, Bio-Rad, against the antigen capture
https://publichealth.jhu.edu/2021/variants- USFDA EUA nucleocapsid (90 min incubation),
vaccines-and-what-they-mean-for-covid- (N) protein visual control
19-testing) Cons: Limited use
by EUA
IgM-CLIA (Datta et al., 2021; https://www. Diazyme DZ-Lite SARS- SARS-CoV-2 Ag Overall: 94.4% Quantitative Pros: High
diazyme.com/covid-19-antibody-tests/dz- CoV-2 IgM CLIA Kit, against the 15–30 days from symptom onset (95% CI): specificity, no
lite-sars-cov-2-igm-clia-kit; https://www. Diazyme Laboratories, nucleocapsid 88.4–97.4 cross-reactivity
fda.gov/media/141255/download) Inc., USFDA EUA (N) and spike Sensitivity of this test after early infection is Cons: Only detects
(S) proteins unknown IgM antibodies
3 COVID-19 diagnostics 101
infected the prokaryote (Ganbaatar & Liu, 2021). The sequences allow the prokary-
ote to detect and destroy DNA from similar bacteriophages during subsequent infec-
tions, protecting the bacterial cell from invasion. Hence, these CRISPR sequences
play a vital role in the antiviral defence of prokaryotes and provide a form of acquired
immunity.
In general, there are two major parts in the CRISPR-Cas system: guide RNA (to
identify and direct Cas endonuclease to the target region) and Cas endonuclease (to
break the target genomic site) (Ding et al., 2020). By delivering the Cas9 nuclease
complexed with a synthetic guide RNA into a cell, the genome can be cleaved at the
desired location, allowing existing genes to be removed and new ones introduced
in vitro or in vivo (in living organisms) (Ganbaatar & Liu, 2021).
enzymatic reporter unlocking (SHERLOCK) can detect clinical sample nucleic acid
sequences in a portable and ultrasensitive manner.
In recent months, Cas12 has been used in several assays. One commonly used
CRISPR test is the DETECTR (SARS-CoV-2 DNA Endonuclease-Targeted
CRISPR Trans Reporter) (Kaminski, Abudayyeh, Gootenberg, Zhang, & Collins,
2021). This assay uses loop-mediated amplification (RT-LAMP) to reverse-
transcribe and amplify viral RNA obtained from a nasal or oral swab. The Cas12
protein then identifies target SARS-CoV-2 sequences, leading to genetic cleavage
that signals a positive test result (Rahimi et al., 2021). The entire test takes only
30–40 min to complete, and the results can be seen using a lateral flow strip. If both
the E and N protein-encoding genes are detected, the DETECTR assay signals a pos-
itive result. However, the result changes to presumptive positive if only one of the
two protein-encoding genes is detected (Rahimi et al., 2021).
The FAM-biotin reporter molecule and lateral flow strips provide a visual result
for the Cas12 detection. Flow strips capture and separate the cleaved strands from the
uncleaved reporter molecules (Rahimi et al., 2021). The RT–LAMP DETECTR
assay’s ability to amplify SARS-CoV-2 nucleic acid directly from the swab sample
is a valuable and critical component of the diagnostic’s efficacy and usability.
Broughton et al. performed a study where they discovered diminishing accuracy
from the diagnostic with higher reaction concentrations of 10% UTM and 10%
phosphate-buffered saline by volume, with estimated LODs decreasing to 15,000
and 500 copies per μL, respectively (Broughton et al., 2020a; Rahimi et al., 2021).
CRISPR-based diagnostic systems have reshaped molecular diagnosis. The
benefits of the CRISPR system, such as speed, precision, specificity, strength, effi-
ciency, and versatility, have inspired researchers to develop CRISPR-based diagnos-
tic and therapeutic methods (Rahimi et al., 2021). For instance, eliminating the need
for thermocycling and isothermal signal amplification offers significant benefits
compared to qRT-PCR, such as fast turnaround time, target specificity for single
nucleotides, integration with usable and user-friendly reporting formats like lateral
flow strips, and no requirement for sophisticated laboratory systems (Rahimi et al.,
2021). During the global COVID-19 outbreak, different groups have begun
designing and developing diagnostic and therapeutic programs based on the
efficient CRISPR system. Thanks to these developments, scientists will be able to
accurately run cost-effective tests and apply these methods to other groundbreaking
molecular work.
and efficient diagnostics for disease detection, and consequently, they have been
harnessed to monitor COVID-19 spread. Sensors for SARS-CoV-2 were designed
based on viral surface nucleoproteins, receptors, and genetic material. Biosensors
can be broadly classified into four categories: (1) optical, (2) thermal, (3) electro-
chemical, and (4) piezoelectric. In the following sections, we discuss the types of
biosensors that are prevalent and their conceptual operation in SARS-CoV-2
diagnostics.
FET Sensor detects SARS-CoV2 antigen protein. Graphene base functionalized 1 fg/mL Seo et al. (2020)
with SARS-CoV2 spike antibody used
Electrochemical SARS-CoV-2 antigen is detected with the potentiostat by measuring the 90 fM within 10–30 s Mahari, Roberts,
change in electrical conductivity. The SARS-CoV2 monoclonal antibody was Shahdeo, and Gandhi
immobilized onto a screen printed carbon electrode (SPCE) (2020)
Electrochemical N and S genes of SARS-CoV2 detected in less than 2 h. RCA is used to 1 copy/ μL Chaibun et al. (2021)
generate amplicons which are hybridized with probes functionalized with
redox active labels
Electrical SARS-CoV2 S1 spike protein detected with the help of Bioelectric Recognition 1 fg/mL Mavrikou,
Assay. The antigen specific antibody is bound to membrane-engineered Moschopoulou,
mammalian cells. As the protein attaches to the antibody considerable change Tsekouras, and Kintzios
is observed in the cellular bioelectric properties (2020)
Electrochemical SARS-CoV2 antibodies are detected by the sensor. Here, the presence of Ab IgG ¼ 0.96 ng/mL Yakoh et al. (2021)
stops a redox conversion which leads to a decrease in current that is IgM ¼ 0.14 ng/mL
measured
Electrochemical RNA of SARS-CoV-2 is detected using calixarene functionalized graphene 200 copies/mL Zhao et al. (2021)
oxide which targets the RNA without any requirement of nucleic acid
amplification and reverse-transcription. Only portable electrochemical
smartphones are a necessity
LFIA (optical) SARS-CoV2 nucleocapsid protein was detected using this platform. Specific 2 ng of antigen Kim et al. (2021)
single chain variable fragment- crystallizable fragment (scfc-fc) fusion protein;
antibodies were developed using phage display technique. Cellulose nano 2.5*104pfu cultured
beads were used for visual display of attached SARS antigen virus
Raman scattering Simultaneous detection of anti SARS-CoV2 IgM/IgG is performed. Dual-layer 1 pg/mL Liu et al. (2021)
based LFIA Raman molecules loaded silver-coated SiO2 nanoparticles as SERS tags in
clinical samples. The Raman molecule used was DTNB; system surface was
modified with CoV2 spike protein which specifically binds antibodies
SPR Dual functional plasmonic biosensor which combines the 0.22 pM Qiu et al. (2020)
plasmonicphotothermal effect and localized surface plasmon resonance
detects the CoV2 specific genes. 2D Au-Nanoislands were functionalized with
complementary DNA receptors which specifically bind to Cov2 sequences
and generate thermoplasmonic heat
SPR SPR sensor developed for the detection of nucleocapsid protein antibodies nM range within Djaileb et al. (2020)
from serum samples. The sensor is coated with the recombinant nucleocapsid 15 min
protein which specifically binds to the antibody from the serum.
Optical Split luciferase mechanism used for antibody detection specific to S protein 89% for S protein; Elledge et al. (2021)
and N protein of SARS-CoV2. NanoLuc was split into two and both arms fused 98% for N protein
with viral antigens. The antibodies bind with both the arms and reconstruct the
NanoLuc, hence generating the luminescence
Continued
Table 4 COVID-19 biosensors designed for rapid detection of disease.—cont’d
Type of sensor Detection LOD or sensitivity Reference
Colorimetric and Detection of N gene of SARS-CoV2 by the use of gold nanoparticles capped 0.18 ng/mL Moitra, Alafeef, Dighe,
SPR with thiol-modified antisense oligonucleotides that are N gene specific. The Frieman, and Pan (2020)
binding of the target demonstrates a change in surface plasmon resonance.
The cleavage of RNA-DNA hybrid gives a detectable precipitate because of
addition of RNase H
Optical (LFIA, Single stranded Recombinase Polymerase Amplification method used for 4 copies/50 μL Kim et al. (2020)
Fluorescence) detection of SARS-CoV2 RNA. Isothermal amplification of RPA is done and sample
then dsDNA converted to a single strand which can be detected using the
hybridization process within 10 min
Optomagnetic Conserved region of SARS-CoV2 RNA dependent RNA polymerase (RdRp) 0.4 fM Tian, Gao, Fock, Dufva,
gene detected by making synthetic cDNA and applying the circle to circle and Hansen (2020)
amplification; a rolling circle amplification based cascade
Fluorescence and E gene and N gene of the SARS-CoV2 are targeted in this assay. Simultaneous 10 copies/μL Broughton et al. (2020b)
LFA reverse transcription and isothermal amplification is done using LAMP. Later
CRISPR technique is used for the detection wherein Cas12 g RNA are utilized
targeting the N and E gene
Fluorescence and CASdetec is a developed nucleic acid detection platform based on CRISPR 1 104 copies/mL Guo et al. (2020)
LFA technique for the detection of RdRp of SARS-CoV2 using sgRNA-3.
Recombinase aided amplification is used to amplify the substrates
Fluorescence and Multiplex reverse transcription loop-mediated isothermal amplification (mRT- 12 copies per reaction Zhu et al. (2020)
LFA LAMP) is designed for the detection of SARS-CoV2 combined with with 100% sensitivity
nanoparticle-based lateral flow biosensor. The opening reading frame 1a/b
and N gene of the virus were detected
Fluorescence, Immunoassay with fluorescence-colorimetry dual mode LFIA for detection of 1:106 dilution by Wang et al. (2020)
colorimetric and IgM and IgG specific to SARS-CoV2 from serum. Spike protein is used for the fluorescence values
LFA detection which is conjugated with SiO2@Au@QD nanobeads
Optical and LFIA Biolayer interferometry technique is used for the detection of spike protein 5 μg/mL Baker et al. (2020)
wherein α,N-acetyl neuraminic acid was immobilized onto sensor platform and
signal was generated upon binding of spike protein to glycan
Optical and LFIA SARS-CoV2 IgG and IgM were detected from serum, plasma and whole 92% sensitivity for Black et al. (2021)
blood. Colloidal-gold labelled SARS-CoV-2 antigen was used for detection venous blood;
93% sensitivity for
capillary blood
Fluorescence and Detection of anti SARS-CoV2 IgG antibody in human serum. The capture – Chen et al. (2020)
LFIA molecule was the recombinant SARS-CoV2 nucleocapsid phosphoprotein.
The detection was done using Lanthanide-doped polystyrene nanoparticles
which served as a fluorescence reporter functionalized with IgG mouse/rabbit
anti-human IgG antibody
LFIA The method developed for simultaneous detection of IgM and IgG specific to Sensitivity ¼ 88.63% Li et al. (2020)
SARS-CoV2. The surface antigen conjugated with gold nanoparticles is used
here as a capture
Optical The sensor is developed based on an interferometric bimodal waveguide Sensitivity ¼ greater Ruiz-Vega, Soler, and
(Interferometry) whose surface is modified with specific receptors targeting antigens of SARS- than 95% Lechuga (2021)
CoV2 such a spike protein. When virus particles are captured by receptors,
interferometric signals are recorded
Plasmonic Sensor Optical detection of SARS-CoV2 virus done using the specific monoclonal 370 virus particles/mL Huang et al. (2021)
antibody and ACE2 protein
LFA Evaluation of four lateral flow assay kits for detection of SARS-CoV2 IgG Sensitivities: McAulay et al. (2020)
BTNX kit 1 ¼ 95%
BTNX kit 2 ¼ 91%
ACON ¼ 95%
SD Biosensor ¼ 92%
LFIA Detection method for SARS-CoV2 spike 1 protein using a combination of 1.86 105 copies/mL Lee et al. (2021)
capture and detector molecule being SARS-CoV2 receptor ACE2 and
commercially available antibodies for S1 respectively
Quartz Crystal S1 spike protein of SARS-CoV2 is detected with the help of anti-spike 40 nM Pandey (2020)
Microbalance glycoprotein (engineered surface with mixed SAM of CH3 and COOH
(Piezo) + antibody)
Amplification and SARS-CoV2 N gene and SARS-CoV2 S gene are being detected using S Xing et al. (2020)
Microfluidic isothermal amplification analyser (RTisochip). The chip is able to detect gene ¼ 10 copies/μL;
device 19 common respiratory viruses as well N
gene ¼ 25 copies/μL
Chromatographic Standard Q COVID-19 antigen (SD Biosensor) for detection of SARS-CoV2 Sensitivity ¼ 70.6% Cerutti et al. (2020)
immunoassay nucleoprotein in nasopharyngeal swabs
108 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
facilitate the electrochemistry of the analyte at the working electrode surface. Poten-
tiometric devices mainly indicate the concentration of ions related to electrochemical
reactions by measuring the charge potential that is accumulated on the working
electrode. Hence, the electromotive force defines the relationship between the con-
centration and the potential at the working electrode, which is also given by the
Nernst equation. Conductometric devices work by measuring the ability of an ana-
lyte to conduct an electrical current between electrodes. They are contemplated as a
subset of impedimetric devices. These devices are widely used for electrochemical
reactions using enzymes, wherein the ionic strength of the conductive solution may
change due to the reaction in situ.
Huge preference for electrochemical transducers is attributed to ease of synthesis,
quick response time, and low power consumption (Table 4) (Desai, Kumar, Bose, &
Datta, 2018; Grieshaber, MacKenzie, V€or€os, & Reimhult, 2008). Seo and his asso-
ciates have developed an electrochemical sensor capable of detecting the S protein
with a sensitivity of 1 fg/mL, whereas Mahari et al. developed a similar sensor with a
sensitivity up to 90 fM (Seo et al., 2020). Later in 2021, Zhao et al. fabricated a tech-
nique for the electrochemical detection of SARS-CoV2 RNA that did not require
nucleic acid amplification (Zhao et al., 2021). Variants and modified electrochemical
sensors have been actively used for COVID19 detection (Table 4).
With face masks becoming a necessity and a preventive safety measure, scientists
have delved into the plausibility of sensor-based face masks as a strategy for iden-
tifying infected individuals. Researchers from Harvard and MIT designed a SARS-
CoV-2 sensing mask capable of detecting a patient within 90 min. Embedded with
miniature sensors, the technology encompasses freeze-dried cellular machinery ca-
pable of detecting viral particles in the exhaled aerosol of the person wearing the
mask. This sensor-based mask has a sensitivity equivalent to that of gold-standard
WHO-approved RT-PCR tests, and it can detect the presence of the viral genome.
This particular novel prototype has begun another path for the next generation sen-
sors. As of now, the prototype biosensor costs approximately $5, and it is anticipated
to become cheaper with mass manufacturing (Nguyen et al., 2021).
Another upcoming and explorative field in next generation sensors are CRISPR-
based sensors. The ability to target several genomic loci simultaneously is achieved
with the help of CRISPR and its associated proteins known as Cas proteins, as men-
tioned previously. With CRISPR being one of the most promising potential fields of
research, the detection of many pathogens are being targeted with the help of
CRISPR, including the detection of coronaviridae. As described before, CRISPR-
based SHERLOCK assays are being utilized specifically for the detection of
SARS-CoV-2. This detection can be achieved by using samples from nasal swabs,
nasopharyngeal swabs, oropharyngeal swabs or bronchoalveolar lavage (BAL). As
the viral signature sequence is detected, the CRISPR enzyme is released to generate
fluorescent signals (Azhar et al., 2020). A CRISPR-based in vitro diagnostic (IVD)
detector is used for the detection of E and N2 genes of SARS-CoV-2 by using
Cas12a. The Cas12a-sgRNA complex binds to the target, leading to ssDNA cleavage
and concurrent increase in fluorescence (Kellner et al., 2019). CRISPR-based detec-
tion offers the user the sensitivity and specificity they require in detection of viruses
belonging to the same family, and it is robust enough to distinguish between genet-
ically similar viruses. This advantage is attributed to the fact that detection is based
on specific signature sequences that are conserved (Datta et al., 2021).
Both viral tests are highly unlikely to result in a false-positive test because of vac-
cination for two primary reasons. First, mRNA COVID-19 vaccines (mRNA-1273
by Moderna and BNT162b2/Comirnaty by Pfizer-BioNTech) and adenovirus-based
COVID-19 vaccines (ChAdOx1-S by AstraZeneca-Oxford and Janssen COVID-19
Vaccine by Johnson & Johnson) do not consist of the entire SARS-CoV-2 genome.
Instead, these four vaccines contain a virus fragment that carries instructions on syn-
thesizing S-proteins. Therefore, these vaccines use the S-protein as a major target.
Fortunately, the majority of widely used molecular tests do not target the S gene.
Moreover, S gene-targeting molecular tests (e.g. Thermofisher Taqpath) have other
gene targets, thereby remaining functionally reliable. Thus, vaccination does not
significantly affect the accuracy of viral tests (Borges et al., 2021).
Second, mRNA-1273 (Moderna) and BNT162b2/Comirnaty (Pfizer-BioNTech)
contain non-replicating mRNA. Specifically, these two vaccines contain the target
antigen sequence, 30 untranslated region (UTR), and 50 UTR but lack protein se-
quences necessary for self-amplification. Consequently, these non-replicating viral
vector vaccines are unable to create new viral particles upon host entry. In other
words, the small amount of mRNA in these non-replicating mRNA vaccines is
too low to be detected by either RT-PCR or antigen testing (Guglielmi, 2020).
On the other hand, vaccination can result in false positive results for serology
(antibody) tests. A false positive can occur if the serology test specifically detects
spike protein-specific antibodies. All four vaccines mentioned in the previous
paragraph (i.e. mRNA-1273, BNT162b2/Comirnaty, ChAdOx1-S, and Janssen
COVID-19 vaccine) aim to elicit antibody production against the spike protein.
Hence, a positive result from serology tests that target the spike protein (Table 3)
could indicate either vaccination or prior infection. In contrast, serology tests that
target antibodies to the nucleocapsid protein (Table 3) provide more conclusive
results; a positive test result could only occur from natural infection, allowing for
differentiation between natural infection and immunization (West, Gronvall, &
Kobokovich, 2021).
Another question is whether serology tests are appropriate diagnostic tools to mea-
sure vaccine-induced immunity against SARS-CoV-2. Unfortunately, there is a lack of
research that evaluates the level of protection provided by an immune response to
COVID-19 vaccination. As of October 2021, the FDA has not recommended any cur-
rently available serology tests as a reliable method to measure vaccination efficacy.
Serology testing remains an unreliable predictor of COVID-19 immunity for two
main reasons. First, scientists and clinicians have yet to determine the exact concen-
tration of antibodies needed to prevent SARS-CoV-2 infection or illness (correlates
of protection). Due to this gap of knowledge, a positive serology test obtained via
qualitative or quantitative COVID-19 serology testing, including those listed in
Table 3, does not guarantee protection against COVID-19. Second, as listed in
Table 3, numerous serology tests target antibodies to the N protein rather than the
S protein. Since currently approved COVID-19 vaccines elicit S protein-specific
neutralizing antibodies, people who have had the COVID-19 vaccine (but not a nat-
ural infection) would test negative with serology tests that do not detect anti-S pro-
tein antibodies. Therefore, serology tests are not an accurate measure of COVID-19
5 Conclusion 111
5 Conclusion
At the time of writing, the COVID-19 pandemic continues, causing widespread in-
fection and death. Diagnostic techniques involving RT-PCR, serology, CRISPR, and
biosensors have all played roles in informing best health practices and slowing the
spread of SARS-CoV-2. However, the disease has become more transmissible,
dangerous, and challenging to detect with the increasing number of emerging vari-
ants. Developments in diagnostic methods have helped, but the spread of the virus
continues to rage. People continue to lose their lives daily, despite the world’s best
diagnostic capabilities. Many countries cannot afford the best diagnostic assays,
resulting in health inequities on a global scale.
It must be mentioned that diagnostics alone are incapable of bringing an end to
the COVID-19 pandemic. While testing helps mitigate the spread, the best way to
stop the spread of COVID-19 is global vaccination, augmenting acquired immunity
against the virus and limiting viral transmission (and severe infection). Vaccination
is crucial to curb variant emergence and transmission frequency while preventing
further viral replication and mutation (Salian et al., 2021). Effective vaccine distri-
bution reduces the need for diagnostic testing since fewer individuals will transmit
and develop viral infections, reducing the population’s viral load. This reduction in
testing demand is vital, given the immense strain that diagnostic efforts have put on
the global scientific and clinical supply chains. Therefore, vaccines are necessary to
stop the spread of COVID-19, mitigate the need for testing, and save lives. Coupled
with implemented public safety measures such as physical distancing, vaccinations
provide hope for a safer and healthier post-pandemic future (Ramos, Vela-Perez,
Ferrández, Kubik, & Ivorra, 2021).
Unfortunately, just as the distribution of testing was not always equal, neither has
the worldwide distribution of vaccines. Many countries have been left out of the ben-
efits of vaccination because they lacked resources and global support (Salian et al.,
2021). Thus, people in these countries must face the dangerous COVID-19 variants
without protection from vaccines. This predictable and sad reality is worsened by
vaccine hesitancy and mistrust within countries with greater vaccine access
(Forni & Mantovani, 2021). These factors contribute to the massive health disparities
related to vaccination worldwide. According to the New York Times, over 5 billion
vaccine doses have been administered worldwide. Yet, billions of people remain
unvaccinated while health disparities worsen. Countries like the United States,
France, China, and Canada have over 100 vaccine doses administered per 100
people. In contrast, countries like Venezuela, Nicaragua, Kenya, and South Africa
average fewer than 20 doses per 100 people (Holder, 2021). These statistics point
to a worldwide issue of access to healthcare that has existed throughout the
COVID-19 pandemic.
112 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
With continued massive testing efforts, valuable resources remain in short sup-
ply, slowing down research and science while infections and deaths persist. The bat-
tle against COVID-19 continues, and a global solution is needed to end it.
A worldwide collaborative effort to continue developing diagnostic assays is crucial
to keeping up with the ever-changing virus and its variants. Researchers and scien-
tists must cooperate and refine diagnostics that are accurate and sensitive to emerg-
ing SARS-CoV-2 variants so new variants can be identified and contained quickly.
Further, these improved diagnostics should be made available to all people: ensuring
the distribution of diagnostic tests to developing countries is a global responsibility.
A globally run task force devoted to monitoring SARS-CoV-2 spread worldwide in
real-time would provide a much-needed solution to the underreported COVID-19
cases around the world. To reiterate an earlier point, equitable vaccine distribution
to people in every nation is also a global responsibility. With a worldwide cooper-
ative effort, future waves of cases caused by new variants can be quickly contained
and ultimately prevented with the proper investment in global accessibility for
COVID-19 testing and vaccination.
References
Abdool Karim, S. S., & de Oliveira, T. (2021). New SARS-CoV-2 Variants—Clinical, public
health, and vaccine implications. The New England Journal of Medicine, 384(19),
1866–1868. https://doi.org/10.1056/NEJMc2100362.
Arevalo-Rodriguez, I., Buitrago-Garcia, D., Simancas-Racines, D., Zambrano-Achig, P.,
Campo, R. D., Ciapponi, A., et al. (2020). False-negative results of initial RT-PCR assays
for COVID-19: A systematic review. PLoS One, 15(12). https://doi.org/10.1371/journal.
pone.0242958, e0242958.
Azhar, M., Phutela, R., Ansari, A. H., Sinha, D., Sharma, N., Kumar, M., et al. (2020). Rapid,
field-deployable nucleobase detection and identification using FnCas9. https://doi.org/
10.1101/2020.04.07.028167.
Babb de Villiers, Chantal, Blackburn, Laura, Cook, Sarah, Janus, Joanna, & PHG Founda-
tion. (2021). SARS-CoV-2 variants. Foundation for Innovative New Diagnostics.
https://www.finddx.org/wp-content/uploads/2021/03/COVID-variants-report-FINAL-
12MAR2021.pdf.
Baker, A. N., Richards, S.-J., Guy, C. S., Congdon, T. R., Hasan, M., Zwetsloot, A. J., et al.
(2020). The SARS-COV-2 spike protein binds sialic acids and enables rapid detection in a
lateral flow point of care diagnostic device. ACS Central Science, 6(11), 2046–2052.
https://doi.org/10.1021/acscentsci.0c00855.
Bausch, D., Hampton, L., Perkins, M., & Saville, M. (2021). COVID-19: Why we can’t use
antibody tests to show that vaccines are working. https://www.gavi.org/vaccineswork/
covid-19-why-we-cant-use-antibody-tests-show-vaccines-are-working.
BioRender.com (2021). https://biorender.com.
Black, M. A., Shen, G., Feng, X., Garcia Beltran, W. F., Feng, Y., Vasudevaraja, V., et al.
(2021). Analytical performance of lateral flow immunoassay for SARS-CoV-2 exposure
screening on venous and capillary blood samples. Journal of Immunological Methods,
489(112909). https://doi.org/10.1016/j.jim.2020.112909.33166549.
References 113
Borges, V., Sousa, C., Menezes, L., Gonçalves, A. M., Picão, M., Almeida, J. P., et al. (2021).
Tracking SARS-CoV-2 lineage B.1.1.7 dissemination: Insights from nationwide spike
gene target failure (SGTF) and spike gene late detection (SGTL) data, Portugal, week
49 2020 to week 3 2021. Eurosurveillance, 26(10). https://doi.org/10.2807/1560-7917.
ES.2021.26.10.2100130.
Broughton, J. P., Deng, X., Yu, G., Fasching, C. L., Servellita, V., Singh, J., et al. (2020a).
CRISPR-Cas12-based detection of SARS-CoV-2. Nature Biotechnology, 38, 870–874.
https://doi.org/10.1038/s41587-020-0513-4.32300245.
Broughton, J. P., Deng, X., Yu, G., Fasching, C. L., Singh, J., Streithorst, J., et al. (2020b).
Rapid detection of 2019 novel coronavirus SARS-CoV-2 using a CRISPR-based
DETECTR lateral flow assay. medRxiv: the preprint server for health sciences. https://
doi.org/10.1101/2020.03.06.20032334. Submitted for publication.
Bustin, S. A., Benes, V., Nolan, T., & Pfaffl, M. W. (2005). Quantitative real-time RT-PCR –
A perspective. Journal of Molecular Endocrinology, 34(3), 597–601. https://doi.org/
10.1677/jme.1.01755.
CDC. (2021, August 7). CDC diagnostic tests for COVID-19. Centers for Disease Control and
Prevention. https://www.cdc.gov/coronavirus/2019-ncov/lab/testing.html.
Cerutti, F., Burdino, E., Milia, M. G., Allice, T., Gregori, G., Bruzzone, B., et al. (2020). Ur-
gent need of rapid tests for SARS CoV-2 antigen detection: Evaluation of the
SD-biosensor antigen test for SARS-CoV-2. Journal of Clinical Virology: The Official
Publication of the Pan American Society for Clinical Virology, 132. https://doi.org/
10.1016/j.jcv.2020.104654, 104654.
Chaibun, T., Puenpa, J., Ngamdee, T., Boonapatcharoen, N., Athamanolap, P., O’Mullane, A.
P., et al. (2021). Rapid electrochemical detection of coronavirus SARS-CoV-2. Nature
Communications, 12(1), 802. https://doi.org/10.1038/s41467-021-21121-7.
Chen, Z., Zhang, Z., Zhai, X., Li, Y., Lin, L., Zhao, H., et al. (2020). Rapid and sensitive de-
tection of anti-SARS-CoV-2 IgG, using lanthanide-doped nanoparticles-based lateral flow
immunoassay. Analytical Chemistry, 92(10), 7226–7231. https://doi.org/10.1021/acs.
analchem.0c00784.
Cheng, L., Song, S., Zhou, B., Ge, X., Yu, J., Zhang, M., et al. (2021). Impact of the N501Y
substitution of SARS-CoV-2 spike on neutralizing monoclonal antibodies targeting di-
verse epitopes. Virology Journal, 18(1), 87. https://doi.org/10.1186/s12985-021-01554-8.
Cherian, S., Potdar, V., Jadhav, S., Yadav, P., Gupta, N., Das, M., et al. (2021). SARS-
CoV-2 spike mutations, L452R, T478K, E484Q and P681R, in the second wave of
COVID-19 in Maharashtra, India. Microorganisms, 9(7), 1542. https://doi.org/10.3390/
microorganisms9071542.
Datta, M., Singh, D. D., & Naqvi, A. R. (2021). Molecular diagnostic tools for the detection of
SARS-CoV-2. International Reviews of Immunology, 40(1–2), 143–156. https://doi.org/
10.1080/08830185.2020.1871477.
Dejnirattisai, W., Zhou, D., Supasa, P., Liu, C., Mentzer, A. J., Ginn, H. M., et al. (2021). An-
tibody evasion by the P.1 strain of SARS-CoV-2. Cell, 184(11). https://doi.org/10.1016/j.
cell.2021.03.055. 2939–2954.e9.
Desai, D., Kumar, A., Bose, D., & Datta, M. (2018). Ultrasensitive sensor for detection of early
stage chronic kidney disease in human. Biosensors & Bioelectronics, 105, 90–94. https://
doi.org/10.1016/j.bios.2018.01.031.
Deyde, V. M., Sampath, R., & Gubareva, L. V. (2011). RT-PCR/electrospray ionization mass
spectrometry approach in detection and characterization of influenza viruses. Expert Re-
view of Molecular Diagnostics, 11(1), 41–52. https://doi.org/10.1586/erm.10.107.
114 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
Di Giacomo, S., Mercatelli, D., Rakhimov, A., & Giorgi, F. M. (2021). Preliminary report on
severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike mutation T478K.
Journal of Medical Virology, 93(9), 5638–5643. https://doi.org/10.1002/jmv.27062.
Ding, X., Yin, K., Li, Z., Lalla, R. V., Ballesteros, E., Sfeir, M. M., et al. (2020). Ultrasensitive
and visual detection of SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nature
Communications, 11(1), 4711. https://doi.org/10.1038/s41467-020-18575-6.
Djaileb, A., Charron, B., Jodaylami, M. H., Thibault, V., Coutu, J., Stevenson, K., et al. (2020).
A rapid and quantitative serum test for SARS-CoV-2 antibodies with portable surface plas-
mon resonance sensing. ChemRxiv. Submitted for publication.
Dyer, O. (2021). Covid-19: Indonesia becomes Asia’s new pandemic epicentre as delta variant
spreads. BMJ, n1815. https://doi.org/10.1136/bmj.n1815.
Elledge, S. K., Zhou, X. X., Byrnes, J. R., Martinko, A. J., Lui, I., Pance, K., et al.
(2021). Engineering luminescent biosensors for point-of-care SARS-CoV-2 antibody
detection. Nature Biotechnology, 39(8), 928–935. https://doi.org/10.1038/s41587-021-
00878-8.
Farasani, A. (2021). Biochemical role of serum ferratin and d-dimer parameters in COVID
19 diagnosis. Saudi Journal of Biological Sciences. https://doi.org/10.1016/j.
sjbs.2021.08.040.
Faria, N. R., Mellan, T. A., Whittaker, C., Claro, I. M., da Candido, D. S., Mishra, S., et al.
(2021). Genomics and epidemiology of the P.1 SARS-CoV-2 lineage in Manaus, Brazil.
Science, 372(6544), 815–821. https://doi.org/10.1126/science.abh2644.
Fomsgaard, A. S., & Rosenstierne, M. W. (2020). An alternative workflow for molecular de-
tection of SARS-CoV-2—Escape from the NA extraction kit-shortage, Copenhagen, Den-
mark, march 2020. Eurosurveillance, 25(14), 2000398. https://doi.org/10.2807/1560-
7917.ES.2020.25.14.2000398.
Forni, G., & Mantovani, A. (2021). COVID-19 vaccines: Where we stand and challenges
ahead. Cell Death and Differentiation, 28(2), 626–639. https://doi.org/10.1038/s41418-
020-00720-9.
Ganbaatar, U., & Liu, C. (2021). CRISPR-based COVID-19 testing: Toward next-generation
point-of-care diagnostics. Frontiers in Cellular and Infection Microbiology, 11, 663949.
https://doi.org/10.3389/fcimb.2021.663949.
Greaney, A. J., Starr, T. N., Barnes, C. O., Weisblum, Y., Schmidt, F., Caskey, M., et al.
(2021). Mutational escape from the polyclonal antibody response to SARS-CoV-2 infec-
tion is largely shaped by a single class of antibodies [preprint]. Microbiology. https://doi.
org/10.1101/2021.03.17.435863.
Green, M. R., & Sambrook, J. (2018). Quantification of RNA by real-time reverse
transcription-polymerase chain reaction (RT-PCR). Cold Spring Harbor Protocols,
2018(10). pdb.prot095042. https://doi.org/10.1101/pdb.prot095042.
Grieshaber, D., MacKenzie, R., V€ or€
os, J., & Reimhult, E. (2008). Electrochemical
biosensors—Sensor principles and architectures. Sensors (Basel, Switzerland), 8(3),
1400–1458. https://doi.org/10.3390/s80314000.
Guglielmi, G. (2020). Fast coronavirus tests: What they can and can’t do. Nature, 585(7826),
496–498. https://doi.org/10.1038/d41586-020-02661-2.
Guo, L., Sun, X., Wang, X., Liang, C., Jiang, H., Gao, Q., et al. (2020). SARS-CoV-2 detection
with CRISPR diagnostics. Cell Discovery, 6(1), 34. https://doi.org/10.1038/s41421-020-
0174-y.
Harvey, W. T., Carabelli, A. M., Jackson, B., Gupta, R. K., Thomson, E. C., Harrison, E. M.,
et al. (2021). SARS-CoV-2 variants, spike mutations and immune escape. Nature Reviews.
Microbiology, 19(7), 409–424. https://doi.org/10.1038/s41579-021-00573-0.
References 115
Heid, C. A., Stevens, J., Livak, K. J., & Williams, P. M. (1996). Real time quantitative PCR.
Genome Research, 6(10), 986–994. https://doi.org/10.1101/gr.6.10.986.
Holder, J. (2021). Covid world vaccination tracker. The New York times. https://www.nytimes.
com/interactive/2021/world/covid-vaccinations-tracker.html.
Huang, L., Ding, L., Zhou, J., Chen, S., Chen, F., Zhao, C., et al. (2021). One-step rapid quan-
tification of SARS-CoV-2 virus particles via low-cost nanoplasmonic sensors in generic
microplate reader and point-of-care device. Biosensors & Bioelectronics, 171, 112685.
https://doi.org/10.1016/j.bios.2020.112685.
Hussain, M., Rupp, F., Wendel, H. P., & Gehring, F. K. (2018). Bioapplications of acoustic
crystals—A review. TrAC Trends in Analytical Chemistry, 102, 194–209. https://doi.org/
10.1016/j.trac.2018.02.009.
Ishikawa, F. N., Chang, H.-K., Curreli, M., Liao, H.-I., Olson, C. A., Chen, P.-C., et al. (2009).
Label-free, electrical detection of the SARS virus N-protein with nanowire biosensors uti-
lizing antibody mimics as capture probes. ACS Nano, 3(5), 1219–1224. https://doi.org/
10.1021/nn900086c.
Jackson, C. B., Zhang, L., Farzan, M., & Choe, H. (2021). Functional importance of the D614G
mutation in the SARS-CoV-2 spike protein. Biochemical and Biophysical Research Com-
munications, 538, 108–115. https://doi.org/10.1016/j.bbrc.2020.11.026.
Kaminski, M. M., Abudayyeh, O. O., Gootenberg, J. S., Zhang, F., & Collins, J. J. (2021).
CRISPR-based diagnostics. Nature Biomedical Engineering, 5(7), 643–656. https://doi.
org/10.1038/s41551-021-00760-7.
Kellner, M. J., Koob, J. G., Gootenberg, J. S., Abudayyeh, O. O., & Zhang, F. (2019). SHER-
LOCK: Nucleic acid detection with CRISPR nucleases. Nature Protocols, 14(10),
2986–3012. https://doi.org/10.1038/s41596-019-0210-2.
Khateeb, J., Li, Y., & Zhang, H. (2021). Emerging SARS-CoV-2 variants of concern and po-
tential intervention approaches. Critical Care, 25(1), 244. https://doi.org/10.1186/s13054-
021-03662-x.
Kim, Y., Yaseen, A. B., Kishi, J. Y., Hong, F., Saka, S. K., Sheng, K., et al. (2020). Single-
strand RPA for rapid and sensitive detection of SARS-CoV-2 RNA. https://doi.org/
10.1101/2020.08.17.20177006.
Kim, H.-Y., Lee, J.-H., Kim, M. J., Park, S. C., Choi, M., Lee, W., et al. (2021). Development
of a SARS-CoV-2-specific biosensor for antigen detection using scFv-fc fusion proteins.
Biosensors & Bioelectronics, 175. https://doi.org/10.1016/j.bios.2020.112868, 112868.
Korber, B., Fischer, W. M., Gnanakaran, S., Yoon, H., Theiler, J., Abfalterer, W., et al. (2020).
Tracking changes in SARS-CoV-2 spike: Evidence that D614G increases infectivity of the
COVID-19 virus. Cell, 182(4). https://doi.org/10.1016/j.cell.2020.06.043. 812–827.e19.
Laffeber, C., de Koning, K., Kanaar, R., & Lebbink, J. H. G. (2021). Experimental evidence for
enhanced receptor binding by rapidly spreading SARS-CoV-2 Variants. Journal of Molec-
ular Biology, 433(15). https://doi.org/10.1016/j.jmb.2021.167058, 167058.
Lee, J.-H., Choi, M., Jung, Y., Lee, S. K., Lee, C.-S., Kim, J., et al. (2021). A novel rapid de-
tection for SARS-CoV-2 spike 1 antigens using human angiotensin converting enzyme 2
(ACE2). Biosensors & Bioelectronics, 171. https://doi.org/10.1016/j.bios.2020.112715,
112715.
Li, Z., Yi, Y., Luo, X., Xiong, N., Liu, Y., Li, S., et al. (2020). Development and clinical ap-
plication of a rapid IgM-IgG combined antibody test for SARS-CoV-2 infection diagnosis.
Journal of Medical Virology, 92(9), 1518–1524. https://doi.org/10.1002/jmv.25727.
Li, B., Deng, A., Li, K., Hu, Y., Li, Z., Xiong, Q., et al. (2021). Viral infection and transmission
in a large, well-traced outbreak caused by the SARS-CoV-2 Delta variant [preprint].
Epidemiology. https://doi.org/10.1101/2021.07.07.21260122.
116 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
Liu, H., Dai, E., Xiao, R., Zhou, Z., Zhang, M., Bai, Z., et al. (2021). Development of a SERS-
based lateral flow immunoassay for rapid and ultra-sensitive detection of anti-SARS-
CoV-2 IgM/IgG in clinical samples. Sensors and Actuators B: Chemical, 329, 129196.
https://doi.org/10.1016/j.snb.2020.129196.
Mahari, S., Roberts, A., Shahdeo, D., & Gandhi, S. (2020). ECovSens-ultrasensitive novel
in-house built printed circuit board based electrochemical device for rapid detection of
nCovid-19 antigen, a spike protein domain 1 of SARS-CoV-2. https://doi.org/
10.1101/2020.04.24.059204.
Martin, J., Klapsa, D., Wilton, T., Zambon, M., Bentley, E., Bujaki, E., et al. (2020). Tracking
SARS-CoV-2 in sewage: Evidence of changes in virus variant predominance during
COVID-19 pandemic. Viruses, 12(10), 1144. https://doi.org/10.3390/v12101144.
Mavrikou, S., Moschopoulou, G., Tsekouras, V., & Kintzios, S. (2020). Development of a por-
table, ultra-rapid and ultra-sensitive cell-based biosensor for the direct detection of the
SARS-COV-2 S1 spike protein antigen. Sensors (Switzerland), 20(11). https://doi.org/
10.3390/s20113121.
McAulay, K., Bryan, A., Greninger, A. L., Grill, F., Lake, D., Kaleta, E. J., et al. (2020).
Retrospective clinical evaluation of 4 lateral flow assays for the detection of SARS-
CoV-2 IgG. Diagnostic Microbiology and Infectious Disease, 98(3), 115161. https://
doi.org/10.1016/j.diagmicrobio.2020.115161.
Meng, B., Kemp, S. A., Papa, G., Datir, R., Ferreira, I. A. T. M., Marelli, S., et al.
(2021). Recurrent emergence of SARS-CoV-2 spike deletion H69/V70 and its role in
the Alpha variant B.1.1.7. Cell Reports, 35(13), 109292. https://doi.org/10.1016/j.
celrep.2021.109292.
Moitra, P., Alafeef, M., Dighe, K., Frieman, M. B., & Pan, D. (2020). Selective naked-eye
detection of SARS-CoV-2 mediated by N gene targeted antisense oligonucleotide
capped Plasmonic nanoparticles. ACS Nano, 14(6), 7617–7627. https://doi.org/10.1021/
acsnano.0c03822.
Morley, A. A. (2014). Digital PCR: A brief history. Biomolecular Detection and Quantifica-
tion, 1(1), 1–2. https://doi.org/10.1016/j.bdq.2014.06.001.
Morris, C. P., Luo, C. H., Amadi, A., Schwartz, M., Gallagher, N., Ray, S. C., et al. (2021). An
update on SARS-CoV-2 diversity in the United States National Capital Region: Evolution
of novel and Variants of concern. Clinical Infectious Diseases: An Official Publication of
the Infectious Diseases Society of America, ciab636. https://doi.org/10.1093/cid/ciab636.
Motozono, C., Toyoda, M., Zahradnik, J., Ikeda, T., Saito, A., Tan, T. S., et al. (2021). An
emerging SARS-CoV-2 mutant evading cellular immunity and increasing viral infectivity
[preprint]. Microbiology. https://doi.org/10.1101/2021.04.02.438288.
Nguyen, P. Q., Soenksen, L. R., Donghia, N. M., Angenent-Mari, N. M., de Puig, H.,
Huang, A., et al. (2021). Wearable materials with embedded synthetic biology sensors
for biomolecule detection. Nature Biotechnology, 1–9. https://doi.org/10.1038/s41587-
021-00950-3.
Nolan, T., Hands, R. E., & Bustin, S. A. (2006). Quantification of mRNA using real-time
RT-PCR. Nature Protocols, 1(3), 1559–1582. https://doi.org/10.1038/nprot.2006.236.
Ong, S. W. X., Chiew, C. J., Ang, L. W., Mak, T.-M., Cui, L., Toh, M. P. H. S., et al. (2021).
Clinical and virological features of SARS-CoV-2 variants of concern: A retrospective co-
hort study comparing B.1.1.7 (Alpha), B.1.315 (Beta), and B.1.617.2 (Delta). Clinical In-
fectious Diseases: An Official Publication of the Infectious Diseases Society of America,
ciab721. https://doi.org/10.1093/cid/ciab721.
References 117
Palaz, F., Kalkan, A. K., Tozluyurt, A., & Ozsoz, M. (2021). CRISPR-based tools: Alternative
methods for the diagnosis of COVID-19. Clinical Biochemistry, 89, 1–13. https://doi.org/
10.1016/j.clinbiochem.2020.12.011.
Pandey, L. M. (2020). Design of engineered surfaces for prospective detection of SARS-CoV-
2 using quartz crystal microbalance-based techniques. Expert Review of Proteomics,
17(6), 425–432. https://doi.org/10.1080/14789450.2020.1794831.
Paweletz, C. P., Charboneau, L., & Liotta, L. A. (2001). Micro RT-PCR. Current Protocols in
Cell Biology, 10(1), A.3G.1–A.3G.6. https://doi.org/10.1002/0471143030.cba03gs10.
Peacock, T. P., Goldhill, D. H., Zhou, J., Baillon, L., Frise, R., Swann, O. C., et al. (2021). The
furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets.
Nature Microbiology, 6(7), 899–909. https://doi.org/10.1038/s41564-021-00908-w.
Peñarrubia, L., Ruiz, M., Porco, R., Rao, S. N., Juanola-Falgarona, M., Manissero, D.,
et al. (2020). Multiple assays in a real-time RT-PCR SARS-CoV-2 panel can mitigate
the risk of loss of sensitivity by new genomic variants during the COVID-19 outbreak.
International Journal of Infectious Diseases, 97, 225–229. https://doi.org/10.1016/
j.ijid.2020.06.027.
Planas, D., Veyer, D., Baidaliuk, A., Staropoli, I., Guivel-Benhassine, F., Rajah, M. M., et al.
(2021). Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Na-
ture, 596(7871), 276–280. https://doi.org/10.1038/s41586-021-03777-9.
Qiu, G., Gai, Z., Tao, Y., Schmitt, J., Kullak-Ublick, G. A., & Wang, J. (2020). Dual-functional
plasmonic photothermal biosensors for highly accurate severe acute respiratory
syndrome coronavirus 2 detection. ACS Nano, 14(5), 5268–5277. https://doi.org/
10.1021/acsnano.0c02439.
Rahimi, H., Salehiabar, M., Barsbay, M., Ghaffarlou, M., Kavetskyy, T., Sharafi, A., et al.
(2021). CRISPR systems for COVID-19 diagnosis. ACS Sensors, 6(4), 1430–1445.
https://doi.org/10.1021/acssensors.0c02312.
Ramos, A. M., Vela-Perez, M., Ferrández, M. R., Kubik, A. B., & Ivorra, B. (2021). Modeling
the impact of SARS-CoV-2 variants and vaccines on the spread of COVID-19. Commu-
nications in Nonlinear Science and Numerical Simulation, 102, 105937. https://doi.org/
10.1016/j.cnsns.2021.105937.
Reardon, S. (2021). How the Delta variant achieves its ultrafast spread. Nature. https://doi.org/
10.1038/d41586-021-01986-w.
Ruiz-Vega, G., Soler, M., & Lechuga, L. M. (2021). Nanophotonic biosensors for point-of-
care COVID-19 diagnostics and coronavirus surveillance. Journal of Physics: Photonics,
3(1), 011002. https://doi.org/10.1088/2515-7647/abd4ee.
Salian, V. S., Wright, J. A., Vedell, P. T., Nair, S., Li, C., Kandimalla, M., et al. (2021).
COVID-19 transmission, current treatment, and future therapeutic strategies. Molecular
Pharmaceutics, 18(3), 754–771. https://doi.org/10.1021/acs.molpharmaceut.0c00608.
Sallam, M., & Mahafzah, A. (2021). Molecular analysis of SARS-CoV-2 genetic lineages in
Jordan: Tracking the introduction and spread of COVID-19 UK variant of concern at a
country level. Pathogens, 10(3), 302. https://doi.org/10.3390/pathogens10030302.
SARS-CoV-2 Total ab assay. (n.d.). Bio-Rad Laboratories. Retrieved August 25, 2021e, from
https://www.bio-rad.com/en-us/product/sars-cov-2-total-ab-assay?ID¼QADMT6E08O1Y.
Seo, G., Lee, G., Kim, M. J., Baek, S.-H., Choi, M., Ku, K. B., et al. (2020). Rapid detection of
COVID-19 causative virus (SARS-CoV-2) in human nasopharyngeal swab specimens
using Field-effect transistor-based biosensor. ACS Nano, 14(4), 5135–5142. https://doi.
org/10.1021/acsnano.0c02823.
118 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
Shahhosseini, N., Babuadze, G. G., Wong, G., & Kobinger, G. P. (2021). Mutation signatures
and in silico docking of novel SARS-CoV-2 variants of concern. Microorganisms, 9(5),
926. https://doi.org/10.3390/microorganisms9050926.
Shrivastav, A. M., Cvelbar, U., & Abdulhalim, I. (2021). A comprehensive review on
plasmonic-based biosensors used in viral diagnostics. Communications Biology, 4(1),
70. https://doi.org/10.1038/s42003-020-01615-8.
Shu, B., Kirby, M. K., Davis, W. G., Warnes, C., Liddell, J., Liu, J., et al. (2021). Multiplex
real-time reverse transcription PCR for influenza A virus, influenza B virus, and severe
acute respiratory syndrome coronavirus 2. Emerging Infectious Diseases Journal—
CDC, 27(7). https://doi.org/10.3201/eid2707.210462.
Shukha, Y., Makhoul, K., Abu-Elhija, J., Hayek, T., & Hamoud, S. (2021). A case of severe
COVID-19 pneumonia diagnosed in bronchoscopy with negative repeated nasopharyngeal
swabs. Journal of Medical Cases, 12(6), 217–219.
Smyrlaki, I., Ekman, M., Lentini, A., Rufino de Sousa, N., Papanicolaou, N., Vondracek, M.,
et al. (2020). Massive and rapid COVID-19 testing is feasible by extraction-free SARS-
CoV-2 RT-PCR. Nature Communications, 11(1), 4812. https://doi.org/10.1038/s41467-
020-18611-5.
Tegally, H., Wilkinson, E., Giovanetti, M., Iranzadeh, A., Fonseca, V., Giandhari, J., et al.
(2021). Detection of a SARS-CoV-2 variant of concern in South Africa. Nature,
592(7854), 438–443. https://doi.org/10.1038/s41586-021-03402-9.
Tian, B., Gao, F., Fock, J., Dufva, M., & Hansen, M. F. (2020). Homogeneous circle-to-circle
amplification for real-time optomagnetic detection of SARS-CoV-2 RdRp coding se-
quence. Biosensors & Bioelectronics, 165, 112356. https://doi.org/10.1016/j.bios.2020.
112356.
Uddin, M., Mustafa, F., Rizvi, T. A., Loney, T., Al Suwaidi, H., Al-Marzouqi, A. H. H., et al.
(2020). SARS-CoV-2/COVID-19: Viral genomics, epidemiology, vaccines, and therapeu-
tic interventions. Viruses, 12(5), 526. https://doi.org/10.3390/v12050526.
Unser, S., Bruzas, I., He, J., & Sagle, L. (2015). Localized surface plasmon resonance
biosensing: Current challenges and approaches. Sensors (Basel, Switzerland), 15(7),
15684–15716. https://doi.org/10.3390/s150715684.
van Dorp, L., Acman, M., Richard, D., Shaw, L. P., Ford, C. E., Ormond, L., et al. (2020).
Emergence of genomic diversity and recurrent mutations in SARS-CoV-2. Infection, Ge-
netics and Evolution, 83. https://doi.org/10.1016/j.meegid.2020.104351, 104351.
Vandenberg, O., Martiny, D., Rochas, O., van Belkum, A., & Kozlakidis, Z. (2020). Consid-
erations for diagnostic COVID-19 tests. Nature Reviews. Microbiology, 1–13. https://doi.
org/10.1038/s41579-020-00461-z.
Vogels, C., Watkins, A. E., Harden, C. A., Brackney, D. E., Shafer, J., Wang, J., et al. (2021).
SalivaDirect: A simplified and flexible platform to enhance SARS-CoV-2 testing capacity.
Med (New York, N.Y.), 2(3), 263–280.e6. https://doi.org/10.1016/j.medj.2020.12.010.
Volz, E., Mishra, S., Chand, M., Barrett, J. C., Johnson, R., Geidelberg, L., et al. (2021). Asses-
sing transmissibility of SARS-CoV-2 lineage B.1.1.7 in England. Nature, 593(7858),
266–269. https://doi.org/10.1038/s41586-021-03470-x.
Wang, C., Yang, X., Gu, B., Liu, H., Zhou, Z., Shi, L., et al. (2020). Sensitive and simultaneous
detection of SARS-CoV-2-specific IgM/IgG using lateral flow immunoassay based on
dual-mode quantum dot nanobeads. Analytical Chemistry, 92(23), 15542–15549.
https://doi.org/10.1021/acs.analchem.0c03484.
References 119
Wang, P., Nair, M. S., Liu, L., Iketani, S., Luo, Y., Guo, Y., et al. (2021). Antibody resistance
of SARS-CoV-2 variants B.1.351 and B.1.1.7. Nature, 593(7857), 130–135. https://doi.
org/10.1038/s41586-021-03398-2.
West, R., Gronvall, G., & Kobokovich, A. (2021). Variants, vaccines and what they mean for
COVID-19 testing. Johns Hopkins Bloomberg School of Public Health. https://www.
jhsph.edu/covid-19/articles/variants-vaccines-and-what-they-mean-for-covid19-testing.
html.
WHO COVID-19 Dashboard. (2020). WHO COVID-19 dashboard. Geneva: World Health
Organization. Available online: Https://covid19.who.int/ (last cited: [Date]). https://
covid19.who.int.
Wolk, D. M., Kaleta, E. J., & Wysocki, V. H. (2012). PCR–electrospray ionization mass
spectrometry. The Journal of Molecular Diagnostics: JMD, 14(4), 295–304. https://doi.
org/10.1016/j.jmoldx.2012.02.005.
Wu, J., & Ju, H. X. (2012). 3.07—Clinical immunoassays and immunosensing. In J. Pawliszyn
(Ed.), Comprehensive sampling and sample preparation (pp. 143–167). Academic Press.
https://doi.org/10.1016/B978-0-12-381373-2.00071-5.
Wu, Y., Xu, W., Zhu, Z., & Xia, X. (2020). Laboratory verification of an RT-PCR assay for
SARS-CoV-2. Journal of Clinical Laboratory Analysis, 34(10), e23507. https://doi.org/
10.1002/jcla.23507.
Xing, W., Liu, Y., Wang, H., Li, S., Lin, Y., Chen, L., et al. (2020). A high-throughput, multi-
index isothermal amplification platform for rapid detection of 19 types of common respi-
ratory viruses including SARS-CoV-2. Engineering (Beijing, China), 6(10), 1130–1140.
https://doi.org/10.1016/j.eng.2020.07.015.
Yakoh, A., Pimpitak, U., Rengpipat, S., Hirankarn, N., Chailapakul, O., & Chaiyo, S. (2021).
Paper-based electrochemical biosensor for diagnosing COVID-19: Detection of
SARS-CoV-2 antibodies and antigen. Biosensors & Bioelectronics, 176, 112912. https://
doi.org/10.1016/j.bios.2020.112912.
Yamamoto, S., Tanaka, A., Kobayashi, S., Oshiro, Y., Ozeki, M., Maeda, K., et al. (2021).
Consistency of the results of rapid serological tests for SARS-CoV-2 among healthcare
workers in a large national hospital in Tokyo, Japan. Global Health & Medicine, 3(2),
90–94. https://doi.org/10.35772/ghm.2021.01022.
Yang, Y., & Du, L. (2021). SARS-CoV-2 spike protein: A key target for eliciting persistent
neutralizing antibodies. Signal Transduction and Targeted Therapy, 6(1), 95. https://doi.
org/10.1038/s41392-021-00523-5.
Zhang, W., Du, R.-H., Li, B., Zheng, X.-S., Yang, X.-L., Hu, B., et al. (2020). Molecular
and serological investigation of 2019-nCoV infected patients: Implication of multiple
shedding routes. Emerging Microbes & Infections, 9(1), 386–389. https://doi.org/
10.1080/22221751.2020.1729071.
Zhao, H., Liu, F., Xie, W., Zhou, T.-C., OuYang, J., Jin, L., et al. (2021). Ultrasensitive
supersandwich-type electrochemical sensor for SARS-CoV-2 from the infected
COVID-19 patients using a smartphone. Sensors and Actuators. B, Chemical, 327,
128899. https://doi.org/10.1016/j.snb.2020.128899.
Zhu, X., Wang, X., Han, L., Chen, T., Wang, L., Li, H., et al. (2020). Multiplex reverse
transcription loop-mediated isothermal amplification combined with nanoparticle-based
lateral flow biosensor for the diagnosis of COVID-19. Biosensors & Bioelectronics,
166, 112437. https://doi.org/10.1016/j.bios.2020.112437.
120 CHAPTER 3 Diagnostics for SARS-CoV-2 and emerging variants
Further reading
Access SARS-CoV-2 IgM Antibody Test. (n.d.). Beckman Coulter. Retrieved August 25, 2021,
from https://www.beckmancoulter.com/products/immunoassay/access-sars-cov-2-igm-an
tibody-test.
ACCESS SARS-CoV-2 IgM. (2020). Beckman Coulter. https://www.fda.gov/media/142911/
download.
Coronavirus (COVID-19) Update: FDA Authorizes First Test that Detects Neutralizing
Antibodies from Recent or Prior SARS-CoV-2 Infection. FDA; FDA. (2020, November 6).
https://www.fda.gov/news-events/press-announcements/coronavirus-covid-19-update-fda-
authorizes-first-test-detects-neutralizing-antibodies-recent-or.
Diazyme DZ-Lite SARS-CoV-2 IgM CLIA Kit. (n.d.). Diazyme Laboratories, Inc. Retrieved
August 25, 2021, from https://www.diazyme.com/covid-19-antibody-tests/dz-lite-sars-
cov-2-igm-clia-kit.
Diazyme Laboratories, Inc. (2020 August 14). DIAZYME DZ-LITE SARS-CoV-2 IgM CLIA
KIT. Diazyme, 1–4. https://www.fda.gov/media/141255/download.
Elecsys® anti-SARS-CoV-2. (n.d.). Diagnostics. Retrieved August 24, 2021b, from https://di
agnostics.roche.com/us/en/products/params/elecsys-anti-sars-cov-2.html.
ELISA: Purpose, Procedure, and Results. (2012, July 19). Healthline. https://www.healthline.
com/health/elisa.
In Vitro Diagnostics EUAs—Molecular Diagnostic Tests for SARS-CoV-2. (2021, August
30). U.S. Food & Drug Administration; FDA. https://www.fda.gov/medical-devices/coro
navirus-disease-2019-covid-19-emergency-use-authorizations-medical-devices/in-vitro-
diagnostics-euas-molecular-diagnostic-tests-sars-cov-2.
Innovita 2019-nCoV Ab Test (Colloidal Gold)—Instructions for Use. (n.d.). https://www.fda.
gov/media/144071/download.
Labcorp’s COVID-19 RT-PCR Test EUA Summary. (2021). 30. https://www.fda.gov/media/
136151/download.
OmniPATH COVID-19 Total Antibody ELISA Test j COVID-19 Antibody Testing. (n.d.).
Thermo Fisher Scientific. Retrieved August 25, 2021, from https://www.thermofisher.
com/covid-19-antibody-testing/us/en/solutions/OmniPATH-COVID19-Total-Antibody-
ELISA-Test.html.
OmniPATH™ COVID-19 Total Antibody ELISA Test. (n.d.). https://www.fda.gov/media/
142700/download.
Platelia SARS-CoV-2 Total Ab. (n.d.). https://www.fda.gov/media/137493/download.
Roche develops new serology test to detect COVID-19 antibodies. (n.d.). Diagnostics. Re-
trieved August 24, 2021d, from shttps://diagnostics.roche.com/global/en/news-listing/
2020/roche-develops-new-serology-test-to-detect-covid-19-antibodies.html.
SalivaDirect Papers & Protocols. (n.d.). Retrieved August 31, 2021, from https://ysph.yale.
edu/salivadirect/publications/.
SARS-CoV-2 Total Antibody assay (CV2T). (n.d.). https://www.fda.gov/media/138757/
download.
Serology: Purpose, Results, and Aftercare. (2012, June 8). Healthline. https://www.healthline.
com/health/serology.
TaqPath COVID-19 multiplex diagnostic solution—US. (n.d.). Retrieved August 31, 2021,
from //www.thermofisher.com/us/en/home/clinical/clinical-genomics/pathogen-detection-
solutions/covid-19-sars-cov-2/multiplex.html.
Further reading 121
TaqPath™ COVID-19 Combo Kit and TaqPath™ COVID-19 Combo Kit Advanced
INSTRUCTIONS FOR USE. (n.d.). https://www.fda.gov/media/136112/download.
Testing Tool Kit, 2021. https://www.centerforhealthsecurity.org/covid-19TestingToolkit/test
ing-basics/types-of-COVID-19-tests/serology-tests.html.
Variants, Vaccines and What They Mean For COVID-19 Testing j Johns Hopkins
Bloomberg School of Public Health. (2021, February 2). Johns Hopkins Bloomberg
School of Public Health. https://publichealth.jhu.edu/2021/variants-vaccines-and-what-
they-mean-for-covid-19-testing.
What is Chemiluminescent immunoassay? j Lorne Laboratories UK. (n.d.). Lorne Laboratories
Ltd. Retrieved August 30, 2021, from https://www.lornelabs.com/news-events/blog/what-
is-chemiluminescent-immunoassay.
This page intentionally left blank
CHAPTER
Abbreviations
AAV adeno-associated vectors
ACE-2 angiotensin converting enzyme-2
AIOD-CRISPR All-In-One Dual CRISPR-Cas12a
CARVER Cas13-assisted restriction of viral expression and readout
Cas CRISPR associated
CASdetect CRISPR-assisted detection
CONAN Cas3-operated nucleic acid detection
COVID-19 Coronavirus Disease 2019
CREST Cas13-based, Rugged, Equitable, Scalable Testing
CRISPR Clustered Regularly Interspaced Short Palindromic Repeat
crRNAs CRISPR RNAs
DETECTR DNA Endonuclease-Targeted CRISPR Trans Reporter
DR direct repeats
EUA emergency use authorization
FELUDA FnCas9 Editor Linked Uniform Detection Assay
GFP green fluorescent protein
gRNA guide RNA
HEDGES High-level Extended Duration Gene Expression System
HEPN higher eukaryotes and prokaryotes nucleotide binding domains
HPV human papilloma virus
IAV influenza A virus
1 Introduction
An outbreak of a new human respiratory disease was noticed in late December 2019
at Wuhan city, Hubei province, China, caused by Severe Acute Respiratory Syn-
drome Coronavirus-2 (SARS-CoV-2), a new emerging corona virus (Jiang et al.,
2020). It has now infected over 211 million people worldwide with over 4.4 million
deaths since its onset (https://covid19.who.int/; accessed on August 24, 2021). The
Global Initiative on Sharing Avian Influenza Data (GISAID) website published the
first genome sequence of SARS-CoV-2 on January 10, 2020 and since then a plethora
of sequences have been released through the GISAID platform (Hu, Guo, Zhou, &
Shi, 2021). On January 9, 2020, the etiological agent was identified as a never seen
before betacoronavirus. Studies claimed that the 2019-nCoV (novel corona virus)
was found to be 96% identical at the whole-genome level to a bat coronavirus
(Zhou et al., 2020). On February 11, 2020, the International Committee on taxonomy
2 Diagnostics and therapeutics for SARS-CoV-2 125
of viruses (ICTV) named the virus 2019-nCoV SARS-CoV-2 and the World Health
Organization (WHO) named the disease coronavirus disease 2019 (COVID-19), then
declared it as a pandemic on March 11, 2020 (https://www.who.int/emergencies/dis
eases/novelcoronavirus2019). The whole of the globe was threatened causing high
morbidity and significant mortality. The SARS-CoV-2 virus belongs to the family
of enveloped positive-sense RNA genome viruses that infects both the upper and
lower respiratory tracts (Lu et al., 2020; Malik et al., 2021). The virus genome is
made up of 06 functional ORFs, arranged (50 to 30 ) as ORF1a/1b (replicase; covers
2/3rd of the 50 genome and encodes for polyprotein 1ab), spike surface glycoprotein
(S), small envelop protein (E), membrane protein (M), and nucleocapsid (N). It also
includes an RNA-dependent RNA polymerase (RdRP) which maintains genome
fidelity (Malik et al., 2021; Sexton et al., 2016).
Among structural proteins, the receptor binding spike surface glycoprotein,
which enables the virus to infect cells, is encoded by the “S gene.” In terms of nu-
cleotide sequence-based analysis, the “S-gene” of SARS-CoV-2 was observed to be
phylogenetically divergent from its previously known counterparts in other corona
viruses (Malik, Kumar, et al., 2020; Malik, Sircar, et al., 2020; Udugama et al.,
2020). However, the receptors used by SARS-CoV-2 are similar to the previously
known SARS-CoV, i.e., angiotensin converting enzyme-2 (ACE-2) (Zhou et al.,
2020). Some of the common symptoms of COVID-19 includes throat pain, fever,
body pain, cough and cold at initial stages of infection. In severe cases there may
be weakness, shortness of breath, skin rashes and congestion of conjunctiva. Certain
cases also revealed difficulty in breathing leading to severe hypoxia causing death
(Malik, Kumar, et al., 2020; Malik, Sircar, et al., 2020).
different methods which helps in early diagnosis of the disease. A list of other adjunct
diagnostic techniques are also exploited/developed to identify the disease including
non-invasive techniques like ultrasound as point-of-care ultrasound (POCUS), CT
scan, piezoelectric biosensing (surface of piezoelectric crystals are bounded by
SARS-CoV2 horse polyclonal antibodies from protein A), gold nanoparticles bio-
sensing (exploited largely for MERS CoV and HCoV viruses), LSPR (Localized sur-
face plasmon resonance approach), and optic immunosensors (LSPCF) (Layqah &
Eissa, 2019). The recorded consistent lung sonographic findings in a case series
of 20 patients in China with confirmed COVID-19 revealed pleural line irregularity
and thickening, focal B-lines, bilateral diffuse B-profile with spared areas, sub-
pleural consolidation and, rare pleural effusion (Peng, Wang, & Zhang, 2020;
Poggiali et al., 2020).
Among all the diagnostic techniques, PCR is one of the most adopted procedures
for detecting viral nucleic acids, and it has been declared as the gold standard
approach to diagnose the viral infections because of high sensitivity and accuracy.
Though, the quantitative PCR (qPCR) may be performed to diagnose COVID-19,
restricted availability to RT-qPCR equipment and materials may cause the diagnos-
tic procedure to take longer and to further complicate the situation because lower
viral loads may go undetected, resulting in false negative results (Broughton
et al., 2020; Wang, Doyle, & Mark, 1989). Hence, there is an immediate need to
adopt a diagnostic method with simplistic, time-efficient and highly accurate
throughput for diagnosing the emerging pathogen in the early stages of infection.
Most recent molecular diagnostics that can detect the presence of infection despite
lower viral titres can be beneficial to ensure timely diagnosis of all infected patients
(Xiang et al., 2020).
A quick detection of the virus serves half the game. However, successful thera-
peutic interventions are equally necessary to combat a viral disease. The develop-
ment of virus specific therapies is a daunting task owing to the involvement of
host factors in viral life cycle, due to which the number of approved antiviral ther-
apies are limited (De Clercq & Li, 2016). The mainstream therapeutic approaches for
the current pandemic of COVID-19 includes preventing the SARS-CoV-2 virus from
multiplication, which can be achieved by the use of the antiviral drugs (antic-
ipated benefits early in the course of disease) and/or with immune modulators,
which can modulate the immune response thereby helping the immune system fight
against the virus (more effective in the later stages of COVID-19) (https://www.
covid19treatmentguidelines.nih.gov/therapeutic-management/). Several hundred
antivirals, immunomodulators, neutralizing antibody therapies etc. are being ana-
lysed to discover effective treatments for the COVID-19 (https://www.raps.org/
news-and-articles/news-articles/2020/3/covid-19-therapeutics-tracker). Due to the
immediate needs, the repurposing of some of the existing antiviral drugs is also seen
as a feasible option. Along with the available options, it is equally important to in-
vestigate the diversified diagnostic and therapeutic approaches. Research is under-
way to develop novel rapid diagnostic techniques and to understand the effect of
distinct categories of potential treatments against SARS-CoV-2.
3 CRISPR/Cas systems 127
In the last few decades, genetic engineering techniques have immensely im-
proved the concept of disease diagnostics and therapeutics. CRISPR biotechnol-
ogies, where the Cas proteins act as effectors to recognize and degrade specific
genome targets, complimentary to a specific guide RNA (primarily a tool for genome
editing), is emerging as a potential tool for the development of new generation
diagnostics, prophylactics as well as therapeutics. In recent years, research has ex-
plored the potential of different CRISPR/Cas systems as a tool for the development
of novel diagnostics and therapeutics, due to specificity, design, feasibility etc. (Cox
et al., 2017).
3 CRISPR/Cas systems
Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR), refers to the
short repeating DNA sequences in the genome of prokaryotes. These sequences were
first identified in E. coli, by Dr. Ishino’s group in 1987, and since then these have
been discovered in different prokaryotes (Mojica, Diez-Villasenor, Soria, & Juez,
2000). When a prokaryote is infected by a virus the repeated sequences are
transcribed into CRISPR RNAs (crRNAs) that guide the CRISPR associated
(Cas) proteins to break the plasmid or viral RNA/DNA sequences. Therefore, the
CRISPR/Cas system is referred to as the adaptive immune system of prokaryotes
(Jinek et al., 2012). The CRISPR/Cas system is classified into class1 and class2,
and has six types (I-VI) with a continuously expanding list of at least 33 subtypes
(Makarova, Wolf, & Koonin, 2018). The hijacking of CRISPR/Cas9 system (class2;
type II) as molecular scissors for genome editing led the royal Swedish academy of
sciences to award the Nobel prize in chemistry (2020) to Emmanuelle Charpentier
and Jenifer A. Doudna (https://www.nobelprize.org/prizes/chemistry/2020/press-
release/). Mechanistically, the CRISPR/Cas systems primarily require two parts; a
nucleic acid binding domain, that recognizes the specific sequence and an effector
protein (Cas) that cleaves/regulates nucleic acids. The basic mechanistic steps in all
the CRISPR/Cas systems described till date are almost the same but the Cas proteins
involved in the processing exhibit substantial diversity. Since its discovery, this sys-
tem has been harnessed for a wide variety of applications, including the development
of novel diagnostic and therapeutic approaches against infectious diseases. In this
chapter, we present the use of different CRISPR/Cas systems for the potential devel-
opment of advanced diagnostic and therapeutic strategies for SARS-CoV-2 4
CRISPR-based diagnostics for SARS-Cov-2.
The CRISPR/Cas systems have an immense capability of transforming the
status of diagnostics and health care systems (Gootenberg et al., 2017) which are
presently ready to take advantage of this technology. The principles of “collateral
cleavage activity” have been exploited by recently developed CRISPR-based
diagnostics in which the developers have made fluorescently labelled ssDNA/
RNA reporter probes to detect the visible bands through the lateral flow assay in
a paper strip, for the development of a novel nucleic acid-based diagnostic tool
128 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
(Chen et al., 2018; Zhang, Abudayyeh, Gootenberg, Sciences, & Mathers, 2020). The
Cas effectors like Cas12a or Cas13 nuclease possessing collateral activity (described in
detail in the next section on therapeutics) are becoming the most popular. Amongst
these the Cas12 effectors are more effective in the detection of tumour associated viral
markers, such as HPV (Chen et al., 2018) and the Cas13 effectors are better at RNA
detection for viruses like Zika and Dengue (Gootenberg et al., 2017). Using this tech-
nology, different Cas proteins are exploited for the development of very efficient di-
agnostic kits for the diagnosis of COVID-19. The various types of Cas systems being
explored for COVID-19 diagnosis are discussed in the next sections of this chapter.
3.1 Cas12
The different approaches developed using various types and sub-types of the Cas12
system are discussed in this section.
3.1.1 Cas12a
Different methods developed using the Cas12a system are discussed in the following
sub-sections and summarized in Table 1.
DETECTR (Broughton Envelop (E) and Uses RT-LAMP for reverse transcription
et al., 2020) Nucleoprotein (N) and isothermal amplification.
Given Emergency Use Authorization
(EUA) by FDA.
AIOD-CRISPR (Ding Nucleoprotein (N) Separate nucleic acid preamplification
et al., 2020; Zhang and multiple manual operations are
et al., 2020) required.
One-pot reaction system with visual
detection.
NER (Wang et al., Orf1a, Orf1b, SARS-CoV-2 nucleic acid detected as
2020) Nucleoprotein green fluorescence under 485 nm light.
(N) and Envelop (E) Portable with high sensitivity and
specificity.
iSCAN (Ali et al., 2020) Envelop (E) and RT-LAMP coupled with
Nucleoprotein (N) CRISPRCas12a.
Suitable for large-scale and early
detection of SARS-CoV-2 carriers.
VaNGuard (Ooi et al., N-gene Highly efficient for detecting viral
2021) mutations.
Robust, time and cost efficient,
sensitive, specific, convenient point-of-
care test for SARS-CoV-2.
3 CRISPR/Cas systems 129
40 min incubation. This assay has been implemented to identify the genomic RNA of
HIV and SARS-CoV-2 with high sensitivity within an hour (Ding, Yin, Li, Lalla,
et al., 2020; Zhang et al., 2020). It was corroborated by testing 28 COVID suspected
clinical swab samples where its ultra- specificity is demonstrated by detection of
HIV-1 with negligible background as compared to the reported real-time RPA
(Safari et al., 2021). The results were found to be consistent with that of the
RT-qPCR method and is developed for rapid, simple, specific, ultrasensitive, one-
pot, and visual detection of SARS-CoV-2. In addition, it replaced the need for a
big incubator with a low-cost hand warmer leading to an instrument-free point of
diagnostic method of COVID-19 (Ding, Yin, Li, Lalla, et al., 2020). A few limita-
tions of the AIOD-CRISPR assay are the need for nucleic acid extraction, and the
need for kits and reagents (Rahimi et al., 2021).
3.1.1.4 iSCAN (in vitro specific CRISPR-based assay for nucleic acids
detection)
The iSCAN system involves the RT-LAMP coupled with CRISPR/Cas12 for the
rapid, specific, accurate, sensitive detection of SARS-CoV2. Its development tar-
geted two regions in N (at the highly conserved 3’end) and E genes wherein the iden-
tified primer set efficiently amplify the synthetic virus fragments, but not the
controls. The LAMP primers were generated to ensure a robust amplification to suf-
fice the LAMP-based detection i.e., 200 bp amplification products. This approach
is specifically suitable for large-scale and early detection of SARS-CoV-2 carriers,
allowing the effective isolation of individuals to limit the spread of the virus.
3 CRISPR/Cas systems 131
Ali et al. validated the detection kit using extracted RNAs from clinical samples of
COVID-19 positive patients (Ali et al., 2020). A requirement of only rapid, field
deployable, simple equipment is highly advantageous in using iSCAN, as the spe-
cific and easy to use RTLAMP and CRISPR/Cas12 takes less than 1 h (colorimetric
reaction coupled to lateral flow immunochromatography makes easy interpretation
of the results). However, the need of kits and reagents remains unsolved (Rahimi
et al., 2021).
3.1.2 Cas12b
Different methods developed using the Cas12b system are discussed in the following
sub-sections and summarized in Table 2.
3.1.3 Cas13
Different methods developed using Cas13 system are discussed in the following
sub-sections and summarized in Table 3.
diagnostic kit for COVID-19 to two targets - one from the S-gene and the other from
the Orf1ab gene. The primers and gRNAs (LwaCas13a CRISPR) were developed
to detect COVID-19 RNA (not cross reacting with related viral genomes) (Hou
et al., 2020). This approach implements a non-targeted reporter RNA tagged to a
fluorescent dye for the identification of specific RNA molecules (Kellner, Koob,
Gootenberg, Abudayyeh, & Zhang, 2019). A web resource containing CRISPR-
Cas13 based assay designs has been developed to identify 67 viruses, including
SARS-CoV-2, Zika virus, and dengue virus, capable of selecting single or multiplex
panels (Chen et al., 2018). The sensitivity of COVID-19 target sequences using
SHERLOCK method is estimated in a range between 10 and 100 copies per micro-
liter of input (20 and 200 aM), i.e., LoD was 10–100 viral RNA copies/μL.
The SHERLOCK COVID-19 detection protocol can be completed in 1 h involv-
ing the following steps:
(1) An isothermal amplification of the sample (25 min incubation) with the help of
recombinase polymerase amplification (RPA) kit.
134 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
(2) Identification of pre-amplified viral RNA with Cas13 (30 min incubation)
(3) Read out of the outcome with paper dipstick (2 min incubation)
SHERLOCK was validated on 154 clinical samples, with 96% and 88% sensitivity
for the fluorescence and lateral flow readouts, respectively (Patchsung et al., 2020).
Additionally, both the assays had 100% specificity (Patchsung et al., 2020). The
advantages of SHERLOCK include amenability to automation and the use of a min-
imum volume of reagents, rapid, sensitive, and no need for sophisticated equipment
(Rahimi et al., 2021).
3.1.4 Cas3
3.1.4.1 Cas3-operated nucleic acid detection (CONAN)
A combination of RT-LAMP and Cas3-based nucleic acid detection, resulted in an
approach known as (CONAN) for COVID-19 diagnosis. Using post RNA isolation
from clinical samples, RT-LAMP was carried out for SARS-CoV-2, and the leading
amplicons were targeted using Cascade/Cas3 to achieve a fluorescence or lateral
flow readout. The lateral flow-based CONAN approach was implemented on 31 clin-
ical samples and showed 90% sensitivity and 95% specificity compared to the
RT-qPCR assay. Similar results were reported with the DETECTR method
(Broughton et al., 2020). Although, Cas3 was implemented for pathogen recognition
for the first time, efficiency was at a level comparable to that of the Cas12a-based
detection method, which has been in use for more than 2 years (Chen et al., 2018).
Therefore, an even more sensitive SARS-CoV-2 detection method can be created by
further optimization of the CONAN method. Advantages of this assay are time and
cost efficiency, highly sensitive, and efficient single-base-pair discrimination.
Whereas, disadvantages are nucleic acid extraction, and need of kits and reagents
(Rahimi et al., 2021).
3.1.5 Cas9
3.1.5.1 FnCas9 editor linked uniform detection assay (FELUDA)
This class of CRISPR/Cas approaches for nucleotide recognition is dependent on the
specific binding and cutting activity of CRISPR/Cas9. Previously, this method was
used for the detection of Zika virus. Explicitly, Cas9 from Francisella novicida
(FnCas9) was found to be highly specific for both target DNA binding and cleavage
(Acharya et al., 2019). Its high specificity helped in developing a FnCas9-based
nucleic acid detection method named (FELUDA) (Azhar et al., 2020), which was
quickly adapted by these authors for diagnosis of COVID-19. This method used
synthetic DNA fragments coding N gene of SARS-CoV-2, demonstrating that this
136 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
Table 4 List of diagnostic methods that can be used for diagnosis of COVID-19.
Method of Cas LODa (limit of
S. No. diagnosis enzyme detection) References
For decades, the biggest hurdle for targeting the viral genome was the lack of precise
gene editing techniques. With the advent of the CRISPR/Cas systems, manipulation
of virus genes has been tested for therapeutic studies in many viruses (Freije et al.,
2019; Lee, 2019). Likewise, CRISPR/Cas systems can be utilized in different ways to
develop anti-SARS-CoV-2 strategies.
4.1.3.2 Cas13b
Like the Cas13a subtype, CRISPR loci encode a single effector protein, Cas13b
which contains two predicted HEPN domains at its N and C- termini. The
CRISPR/Cas13b system cleaves ssRNA using HEPN domains and also exhibits col-
lateral RNase activity. Apart from HEPN domains there is no sequence similarity
between 13a and 13b. Unlike Cas13a, Cas1 and Cas2 are absent in the Cas13b system
and it can process its own CRISPR array. The Cas13b system has two variants
denoted as VI-B1 and VI-B2 with their accessory proteins named as Csx27 and
Csx28 respectively. In the VI-B1 system, the Csx27 causes repression of Cas13b
whereas Csx28 enhances Cas13b activity. It has been shown that along with several
factors, the presence of secondary structure in the RNA target impacts the activity of
Cas13b enzymes (Smargon et al., 2017). A research group from Thailand showed
that CRISPR/Cas13b system effectively abrogated the porcine reproductive and
respiratory syndrome virus (PRRSV), by simultaneously targeting ORF5 and
ORF7 genes (Cui, Techakriengkrai, Nedumpun, & Suradhat, 2020).
The antiviral potential of Cas13 systems (both 13a and 13b) have been evaluated
in 3 different ssRNA viruses [lymphocytic choriomeningitis virus, (LCMV), Influ-
enza A virus (IAV) and vesicular stomatitis virus (VSV)] in cell culture by Freije
et al. (2019). They carried out a computational analysis followed by experimental
validation of viral genome sequences to create a repository of antiviral crRNAs.
They used Cas13a (LwaCas13a) and Cas13b (PspCas13b) from L. wadei and Pre-
votella sp. P5–125, respectively, to demonstrate the generalizability of the Cas13
system. Their study demonstrated that Cas13 can efficiently reduce the viral RNA
levels in mammalian cells, confirming the potent antiviral activity of Cas13 against
different ssRNA viruses (Freije et al., 2019). In comparison to the Cas13a system,
140 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
the CRISPR/Cas13b system was found to be more robust and its activity can be
upregulated or downregulated depending on the accessory proteins encoded by its
loci (Cox et al., 2017). Considering that SARS-CoV-2 also possess a ssRNA viral
genome, the same principles can be applied to generate anti SARS-CoV-2
therapeutics.
4.1.3.3 Cas13d
For working with this system, guide RNA is made up of a 30 nt direct repeat with a
22 nt spacer (Fig. 1). Spacer length of 22 nt is desirable as below this length the cleav-
age activity is significantly dropped. In mammalian cells, the collateral cleavage
activity of Cas13d is absent. Also, the target cleavage does not depend on the flank-
ing sequence requirements but the cleavage pattern varies with the target. For exam-
ple, Eubacterium siraeum/EsCas13d prefers uracil bases in the target region above
all other bases. The study was conducted to find out an active Cas13d orthologue in
eukaryotic cells and revealed that an engineered variant of Cas13d Ruminococcus
flavefaciens strain XPD3002 (Rfx) Cas13d (CasRx) can be developed into a flexible
tool for programmable ssRNA targeting in mammalian cells. They compared the
small hairpin RNA (shRNA) interference, CRISPR subtype VI-A/VI-B and Cas9
mediated transcriptional inhibition (CRISPRi) with CasRx and found that CasRx
outperformed with 96% knockdown as compared to 65% knockdown by shRNA, ap-
proximately 70% by CRISPRi and 53% by CRISPRi; suggesting CasRx was the most
efficient RNA regulating method. Another study by Yan and co-workers, character-
ized E. siraeum (EsCas13d) and Ruminococcus sp. (RspCas13d) orthologues of
Cas13d enzyme (Yan et al., 2018). They also showed that Cas13d associated acces-
sory proteins have a WYL domain, because the target activity of RspCas13d was
increased, indicating that this particular domain regulates the activity of Cas13d
(Yan et al., 2018). Recently, a research group from Stanford University developed
a potential CRISPR/Cas13d-based pan-coronavirus inhibition strategy. They named
it prophylactic antiviral CRISPR in human cells (PAC-MAN) and showed that it
could cleave the SARS-CoV-2 sequences that were effective as novel antiviral ther-
apy against COVID-19 (Abbott et al., 2020). Abbott and colleagues used CasRx and
synthesized 20 crRNAs targeting RNA-dependent RNA polymerase (RdRp) and Nu-
cleocapsid (N) genes, each. They chose a Cas13d expressing human lung epithelial
cell line (A549) to transduce the pool of crRNAs and found that the targeting RdRp
and N gene repressed a reporter by 86% and 71% respectively. It was also demon-
strated that the SARS-CoV-2 inhibition was quite sensitive to different crRNA con-
centrations and lowering the Cas13d expression has little effect on the inhibitory
activity of the Cas13d system (Abbott et al., 2020). They also suggested that
CRISPR/Cas13d can be used as an antiviral strategy, both prophylactically and ther-
apeutically (Fig. 2).
Consolidating the studies on antiviral capabilities of subtypes of CRISPR/Cas13
systems, Cox and co-workers detected that among the different orthologues of
Cas13a, b and c; Cas13b (PspCas13b) was the most specific and efficient for
RNA knockdown in mammalian cells (Cox et al., 2017). LwaCas13a had two major
FIG. 1
Schematics for SARS-Cov-2 detection using CRISPR/Cas platform. The RNA is extracted from the patient using conventional RNA extraction
method. For Cas12 based detection the RNA is amplified into dsDNA whereas for Cas13 based detection the amplified DNA is transcribed into
ssRNA. The collateral activity of both Cas12 and Cas13 is used for the detection of SARS-CoV-2.
142 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
FIG. 2
(A) CRISPR/Cas13d array showing HEPN domains. (B) Schematic of CRISPR/Cas13d
mechanism as an antiviral strategy. Cas13d shown as a green cloud and guide RNA (DR and
spacer) together forms a Cas13d:crRNA complex required for viral RNA degradation. DR:
Direct repeat.
issues; first it must be stabilized with monomeric super-folded GFP and secondly, the
average RNA knockdown efficiency was around 50% whereas PspCas13b provided
better efficiency as compared to LwaCas13a with 62.9% knockdown. Although
these systems can be reprogrammed to target ssRNA, it is difficult to pack them into
adeno-associated vectors (AAV) for in-vivo delivery due to their large size. In
comparison to Cas13a (1250 aa), Cas13b (1150 aa), and Cas13c (1120 aa), Cas13d
effector has an average length of 930 aa (the smallest class2 CRISPR effector) (Cox
et al., 2017; Shmakov et al., 2015; Smargon et al., 2017). Overall, the CRISPR/
Cas13d system seems to be the most practical system for a therapeutic approach
to target the SARS-CoV-2 genome. The small but robust CRISPR/Cas13d system
seems to be the latest technology for the RNA engineering toolbox.
This claim may be further supported by the following statements.
i. The size of Cas13d (approx. 930aa) is 17–26% smaller than other class2 type VI-
CRISPR/Cas subtypes (Cas13a-c); which makes it suitable for packaging in
low- capacity vectors, like AAV; making it particularly suitable for delivery
in primary cells and in-vivo applications.
ii. It lacks any sequence constraints for flanking sequences i.e., Cas13d does not
require the presence of PFS. This property makes it possible to target
theoretically any ssRNA sequence.
4 CRISPR-based therapeutics for SARS-Cov-2 143
Therefore, the strong catalytic activity, high specificity, and small size of the
Cas13d protein makes it the best choice for targeting the SARS-CoV-2 genome
(Abbott et al., 2020). Studies showed that the highly conserved regions of SARS-
CoV-2 [(Abbott et al., 2020) nucleocapsid (N) which protects the viral genome
and (Abudayyeh et al., 2017) RNA-dependent RNA polymerase (RdRp) which ca-
talyses the transcription of all viral mRNAs] can become potent targets of CRISPR/
Cas13d (Chan et al., 2020), thus disabling virus production and function (Abbott
et al., 2020).
i. First and foremost is the lack of a safe and effective in-vivo delivery method into
human respiratory tract cells: As the Cas13d enzyme has a small size, adeno-
associated virus (AAV) seems to be the most feasible option but the adaptive
immune response against AAV might be of a concern. Hopefully, one of the
methods mentioned in Section 6 could be used for the CRISPR based antiviral
delivery in humans using a nasal spray/nebulizer system, in the future.
ii. Off-target effects: An evaluation of the off-target activity of the crRNAs using
whole transcriptome RNA sequencing would be required.
7 Summary 145
7 Summary
Since the beginning of the COVID-19 pandemic, many diagnostic approaches
(RT-qPCR, RAPID, LFA) have been adopted, among which RT-qPCR turned out
to be the most popular/gold standard. But, one of the mystifying facets of
COVID-19 is its presentation of a wide range of symptoms which varies among dif-
ferent patients and needs early diagnosis for better management. Even though
RT-qPCR is a precise molecular technique false negative results may be obtained.
On the other hand, CRISPR-based SARS-CoV-2 detection approaches are cost
and time efficient, highly sensitive and specific, and do not require sophisticated
instruments. Moreover, they also have shown promise to increase scalability and
enable the diagnostic tests to be carried out at the point-of-care (POC). The CRISPR
can be customized to the target for any genomic region of interest within the desired
genome possessing a broad range of other applications and has been efficiently
implemented for diagnosis of SARS-CoV-2. Considering the therapeutics, we need
to understand that the traditional vaccines priming the immune system against the
exposure to the viral proteins shows a high rate of mutations, overcoming the host
immune response. In comparison, antiviral therapies targeting the highly conserved
gene sequences can limit the escape of SARS-CoV-2 by the immune system. The
CRISPR/Cas systems provide specific gene targeting with immense potential to
develop new generation diagnostics and therapeutics. Moreover, with the
CRISPR/Cas based therapeutics, multiplexing is possible, where different sgRNAs
or crRNAs can be guided to more than one target within the same gene thus decreas-
ing the possibility of viral escape mutants. As an exceptionally efficient tool the
CRISPR/Cas13 system, CARVER (Cas13-assisted restriction of viral expression
and readout) can be implemented to target a broad range of ssRNA viruses, and it
can be used for both, diagnosis and treatment for a variety of viral diseases including
SARS-CoV-2. However, the efficacy and safety of the CRISPR-based therapeutics
needs to be assessed in pre-clinical and clinical settings. Although the CRISPR bio-
technologies are not very helpful to control the present pandemic of COVID-19 there
is hope that the limitations of the CRISPR/Cas system can be overcome in the near
future. The CRISPR based strategies would lead to a new era in the field of disease
diagnosis and therapeutic development, that would make us better prepared for
future viral threats.
146 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
References
Abbott, T. R., Dhamdhere, G., Liu, Y., Lin, X., Goudy, L., Zeng, L., et al. (2020). Develop-
ment of CRISPR as an antiviral strategy to combat SARS-CoV-2 and influenza. Cell,
181(4), 865–876. e812. https://doi.org/10.1016/j.cell.2020.04.020.
Abudayyeh, O. O., Gootenberg, J. S., Essletzbichler, P., Han, S., Joung, J., Belanto, J. J., et al.
(2017). RNA targeting with CRISPR-Cas13. Nature, 550(7675), 280–284. https://doi.org/
10.1038/nature24049.
Abudayyeh, O. O., Gootenberg, J. S., Konermann, S., Joung, J., Slaymaker, I. M., Cox, D. B.,
et al. (2016). C2c2 is a single-component programmable RNA-guided RNA-targeting
CRISPR effector. Science, 353(6299), aaf5573. https://doi.org/10.1126/science.aaf5573.
Acharya, S., Mishra, A., Paul, D., Ansari, A. H., Azhar, M., Kumar, M., et al. (2019).
Francisella novicida Cas 9 interrogates genomic DNA with very high specificity and
can be used for mammalian genome editing. Proceedings of the National Academy of Sci-
ences of the United States of America, 116(42), 20959–20968. https://doi.org/10.1073/
pnas.1818461116.
Ali, Z., Aman, R., Mahas, A., Rao, G. S., Tehseen, M., Marsic, T., et al. (2020). iSCAN: An
RT-LAMP-coupled CRISPR-Cas12 module for rapid, sensitive detection of SARS-CoV-
2. Virus Research, 288, 198129. https://doi.org/10.1016/j.virusres.2020.198129.
Amirkhanov, R., & Stepanov, G. (2019). Systems of delivery of CRISPR/Cas9 ribonucleopro-
tein complexes for genome editing. Russian Journal of Bioorganic Chemistry, 45(6),
431–437.
Azhar, M., Phutela, R., Ansari, A. H., Sinha, D., Sharma, N., Kumar, M., et al. (2020). Rapid,
field-deployable nucleobase detection and identification using FnCas9. bioRxiv,
2020.2004.2007.028167. https://doi.org/10.1101/2020.04.07.028167.
Blanchard, E. L., Vanover, D., Bawage, S. S., Tiwari, P. M., Rotolo, L., Beyersdorf, J., et al.
(2021). Treatment of influenza and SARS-CoV-2 infections via mRNA-encoded Cas13a
in rodents. Nature Biotechnology, 39(6), 717–726. https://doi.org/10.1038/s41587-021-
00822-w.
Brandsma, E., Verhagen, H., van de Laar, T. J. W., Claas, E. C. J., Cornelissen, M., & van den
Akker, E. (2020). Rapid, sensitive and specific SARS coronavirus-2 detection: A multi-
center comparison between standard qRT-PCR and CRISPR based DETECTR. Journal
of Infectious Diseases. https://doi.org/10.1093/infdis/jiaa641.
Broughton, J. P., Deng, X., Yu, G., Fasching, C. L., Servellita, V., Singh, J., et al. (2020).
CRISPR-Cas12-based detection of SARS-CoV-2. Nature Biotechnology, 38(7), 870–874.
https://doi.org/10.1038/s41587-020-0513-4.
Burmistrz, M., Krakowski, K., & Krawczyk-Balska, A. (2020). RNA-targeting CRISPR-Cas
systems and their applications. International Journal of Molecular Sciences, 21(3). https://
doi.org/10.3390/ijms21031122.
Chan, J. F., Kok, K. H., Zhu, Z., Chu, H., To, K. K., Yuan, S., et al. (2020). Genomic char-
acterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with
atypical pneumonia after visiting Wuhan. Emerging Microbes & Infections, 9(1),
221–236. https://doi.org/10.1080/22221751.2020.1719902.
Chen, J. S., Ma, E., Harrington, L. B., Da Costa, M., Tian, X., Palefsky, J. M., et al. (2018).
CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity.
Science, 360(6387), 436–439. https://doi.org/10.1126/science.aar6245.
References 147
Cox, D. B. T., Gootenberg, J. S., Abudayyeh, O. O., Franklin, B., Kellner, M. J., Joung, J., et al.
(2017). RNA editing with CRISPR-Cas13. Science, 358(6366), 1019–1027. https://doi.
org/10.1126/science.aaq0180.
Cui, J., Techakriengkrai, N., Nedumpun, T., & Suradhat, S. (2020). Abrogation of PRRSV
infectivity by CRISPR-Cas13b-mediated viral RNA cleavage in mammalian cells. Scien-
tific Reports, 10(1), 9617. https://doi.org/10.1038/s41598-020-66775-3.
Daniloski, Z., Jordan, T. X., Wessels, H. H., Hoagland, D. A., Kasela, S., Legut, M., et al.
(2021). Identification of required host factors for SARS-CoV-2 infection in human cells.
Cell, 184(1), 92–105. e116. https://doi.org/10.1016/j.cell.2020.10.030.
De Clercq, E., & Li, G. (2016). Approved antiviral drugs over the past 50 years. Clinical
Microbiology Reviews, 29(3), 695–747. https://doi.org/10.1128/cmr.00102-15.
Ding, X., Yin, K., Li, Z., Lalla, R. V., Ballesteros, E., Sfeir, M. M., et al. (2020). Ultrasensitive
and visual detection of SARS-CoV-2 using all-in-one dual CRISPR-Cas12a assay. Nature
Communications, 11(1), 4711. https://doi.org/10.1038/s41467-020-18575-6.
East-Seletsky, A., O’Connell, M. R., Knight, S. C., Burstein, D., Cate, J. H., Tjian, R., et al.
(2016). Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and
RNA detection. Nature, 538(7624), 270–273. https://doi.org/10.1038/nature19802.
East-Seletsky, A., O’Connell, M. R., Burstein, D., Knott, G. J., & Doudna, J. A. (2017). RNA
targeting by functionally orthogonal type VI-A CRISPR-Cas enzymes. Molecular Cell,
66(3), 373–383. e373. https://doi.org/10.1016/j.molcel.2017.04.008.
Freije, C. A., Myhrvold, C., Boehm, C. K., Lin, A. E., Welch, N. L., Carter, A., et al. (2019).
Programmable inhibition and detection of RNA Viruses using Cas13. Molecular Cell,
76(5), 826–837. e811. https://doi.org/10.1016/j.molcel.2019.09.013.
Gao, Z., Fan, M., Das, A. T., Herrera-Carrillo, E., & Berkhout, B. (2020). Extinction of all
infectious HIV in cell culture by the CRISPR-Cas12a system with only a single crRNA.
Nucleic Acids Research, 48(10), 5527–5539. https://doi.org/10.1093/nar/gkaa226.
Gootenberg, J. S., Abudayyeh, O. O., Lee, J. W., Essletzbichler, P., Dy, A. J., Joung, J., et al.
(2017). Nucleic acid detection with CRISPR-Cas13a/C2c2. Science, 356(6336), 438–442.
https://doi.org/10.1126/science.aam9321.
Gootenberg, J. S., Abudayyeh, O. O., Kellner, M. J., Joung, J., Collins, J. J., & Zhang, F.
(2018). Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a,
and Csm6. Science, 360(6387), 439–444. https://doi.org/10.1126/science.aaq0179.
Guan, S., Munder, A., Hedtfeld, S., Braubach, P., Glage, S., Zhang, L., et al. (2019). Self-
assembled peptide-poloxamine nanoparticles enable in vitro and in vivo genome restora-
tion for cystic fibrosis. Nature Nanotechnology, 14(3), 287–297. https://doi.org/10.1038/
s41565-018-0358-x.
Guo, L., Sun, X., Wang, X., Liang, C., Jiang, H., Gao, Q., et al. (2020). SARS-CoV-2 detection with
CRISPR diagnostics. Cell Discovery, 6(1), 34. https://doi.org/10.1038/s41421-020-0174-y.
Handumrongkul, C., Ye, A. L., Chmura, S. A., Soroceanu, L., Mack, M., Ice, R. J., et al.
(2019). Durable multitransgene expression in vivo using systemic, nonviral DNA delivery.
Science Advances, 5(11), eaax0217. https://doi.org/10.1126/sciadv.aax0217.
Hou, T., Zeng, W., Yang, M., Chen, W., Ren, L., Ai, J., et al. (2020). Development and
evaluation of a rapid CRISPR-based diagnostic for COVID-19. PLoS Pathogens, 16(8),
e1008705. https://doi.org/10.1371/journal.ppat.1008705.
Hu, B., Guo, H., Zhou, P., & Shi, Z. L. (2021). Characteristics of SARS-CoV-2 and COVID-
19. Nature Reviews. Microbiology, 19(3), 141–154. https://doi.org/10.1038/s41579-020-
00459-7.
148 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
Jiang, S., Shi, Z., Shu, Y., Song, J., Gao, G. F., Tan, W., et al. (2020). A distinct name is needed
for the new coronavirus. Lancet, 395(10228), 949. https://doi.org/10.1016/s0140-6736
(20)30419-0.
Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A., & Charpentier, E. (2012).
A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.
Science, 337(6096), 816–821. https://doi.org/10.1126/science.1225829.
Joung, J., Ladha, A., Saito, M., Segel, M., Bruneau, R., Huang, M. W., et al. (2020). Point-of-
care testing for COVID-19 using SHERLOCK diagnostics. medRxiv. https://doi.org/
10.1101/2020.05.04.20091231.
Kellner, M. J., Koob, J. G., Gootenberg, J. S., Abudayyeh, O. O., & Zhang, F. (2019). SHER-
LOCK: Nucleic acid detection with CRISPR nucleases. Nature Protocols, 14(10),
2986–3012. https://doi.org/10.1038/s41596-019-0210-2.
Krishnamurthy, S., Wohlford-Lenane, C., Kandimalla, S., Sartre, G., Meyerholz, D. K.,
Theberge, V., et al. (2019). Engineered amphiphilic peptides enable delivery of proteins
and CRISPR-associated nucleases to airway epithelia. Nature Communications, 10(1),
4906. https://doi.org/10.1038/s41467-019-12922-y.
Layqah, L. A., & Eissa, S. (2019). An electrochemical immunosensor for the corona virus as-
sociated with the Middle East respiratory syndrome using an array of gold nanoparticle-
modified carbon electrodes. Mikrochimica Acta, 186(4), 224. https://doi.org/10.1007/
s00604-019-3345-5.
Lee, C. (2019). CRISPR/Cas9-based antiviral strategy: Current status and the potential chal-
lenge. Molecules, 24(7). https://doi.org/10.3390/molecules24071349.
Lin, X., Liu, Y., Chemparathy, A., Pande, T., La Russa, M., & Qi, L. S. (2021).
A comprehensive analysis and resource to use CRISPR-Cas13 for broad-spectrum
targeting of RNA viruses. Cell Reports Medicine, 2(4), 100245. https://doi.org/10.1016/
j.xcrm.2021.100245.
Lu, R., Zhao, X., Li, J., Niu, P., Yang, B., Wu, H., et al. (2020). Genomic characterisation and
epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor
binding. Lancet, 395(10224), 565–574. https://doi.org/10.1016/s0140-6736(20)30251-8.
Mahase, E. (2020). Covid-19: Coronavirus was first described in the BMJ in 1965. BMJ, 369,
m1547. https://doi.org/10.1136/bmj.m1547.
Makarova, K. S., Wolf, Y. I., & Koonin, E. V. (2018). Classification and nomenclature of
CRISPR-Cas systems: Where from here, The CRISPR Journal, 1(5), 325–336, Published
online 2018 Oct 17. https://doi.org10.1089/crispr.2018.0033.
Malik, Y. S., Sircar, S., Bhat, S., Ansari, M. I., Pande, T., Kumar, P., et al. (2020). How
artificial intelligence may help the Covid-19 pandemic: Pitfalls and lessons for the future.
Reviews in Medical Virology, e2205. https://doi.org/10.1002/rmv.2205.
Malik, Y. S., Kumar, N., Sircar, S., Kaushik, R., Bhat, S., Dhama, K., et al. (2020). Corona-
virus disease pandemic (COVID-19): Challenges and a global perspective. Pathogens,
9(7). https://doi.org/10.3390/pathogens9070519.
Malik, Y. S., Ansari, M. I., Kattoor, J. J., Kaushik, R., Sircar, S., Subbaiyan, A., et al. (2021).
Evolutionary and codon usage preference insights into spike glycoprotein of SARS-CoV-
2. Briefings in Bioinformatics, 22(2), 1006–1022. https://doi.org/10.1093/bib/bbaa383.
Marraffini, L. A., & Sontheimer, E. J. (2010). Self versus non-self discrimination during
CRISPR RNA-directed immunity. Nature, 463(7280), 568–571. https://doi.org/10.1038/
nature08703.
Mojica, F. J. M., Diez-Villasenor, C., Soria, E., & Juez, G. (2000). Biological significance of a
family of regularly spaced repeats in the genomes of Archaea, Bacteria and mitochondria.
Molecular Microbiology, 36, 244–246.
References 149
Myhrvold, C., Freije, C. A., Gootenberg, J. S., Abudayyeh, O. O., Metsky, H. C., Durbin, A. F.,
et al. (2018). Field-deployable viral diagnostics using CRISPR-Cas13. Science, 360(6387),
444–448. https://doi.org/10.1126/science.aas8836.
O’Connell, M. R. (2019). Molecular mechanisms of RNA targeting by Cas13-containing type
VI CRISPR-Cas systems. Journal of Molecular Biology, 431(1), 66–87. https://doi.org/
10.1016/j.jmb.2018.06.029.
Ooi, K. H., Liu, M. M., Tay, J. W. D., Teo, S. Y., Kaewsapsak, P., Jin, S., et al. (2021). An
engineered CRISPR-Cas12a variant and DNA-RNA hybrid guides enable robust and rapid
COVID-19 testing. Nature Communications, 12(1), 1739. https://doi.org/10.1038/s41467-
021-21996-6.
Patchsung, M., Jantarug, K., Pattama, A., Aphicho, K., Suraritdechachai, S., Meesawat, P.,
et al. (2020). Clinical validation of a Cas13-based assay for the detection of SARS-
CoV-2 RNA. Nature Biomedical Engineering, 4(12), 1140–1149. https://doi.org/
10.1038/s41551-020-00603-x.
Peng, Q. Y., Wang, X. T., & Zhang, L. N. (2020). Findings of lung ultrasonography of novel
corona virus pneumonia during the 2019-2020 epidemic. Intensive Care Medicine, 46(5),
849–850. https://doi.org/10.1007/s00134-020-05996-6.
Pizzol, J. L. D., Hora, V. P. D., Reis, A. J., Vianna, J., Ramis, I., Groll, A. V., et al. (2020).
Laboratory diagnosis for Covid-19: A mini-review. Revista da Sociedade Brasileira de
Medicina Tropical, 53, e20200451. https://doi.org/10.1590/0037-8682-0451-2020.
Poggiali, E., Dacrema, A., Bastoni, D., Tinelli, V., Demichele, E., Mateo Ramos, P.,
et al. (2020). Can lung US help critical care clinicians in the early diagnosis of novel
coronavirus (COVID-19) pneumonia? Radiology, 295(3), E6. https://doi.org/10.1148/radiol.
2020200847.
Rahimi, H., Salehiabar, M., Barsbay, M., Ghaffarlou, M., Kavetskyy, T., Sharafi, A., et al.
(2021). CRISPR systems for COVID-19 diagnosis. ACS Sensors, 6(4), 1430–1445.
https://doi.org/10.1021/acssensors.0c02312.
Ramachandran, A., Huyke, D. A., Sharma, E., Sahoo, M. K., Huang, C., Banaei, N., et al.
(2020). Electric field-driven microfluidics for rapid CRISPR-based diagnostics and its
application to detection of SARS-CoV-2. Proceedings of the National Academy of Sci-
ences of the United States of America, 117(47), 29518–29525. https://doi.org/10.1073/
pnas.2010254117.
Rauch, J. N., Valois, E., Solley, S. C., Braig, F., Lach, R. S., Audouard, M., et al. (2021).
A scalable, easy-to-deploy protocol for Cas13-based detection of SARS-CoV-2 genetic
material. Journal of Clinical Microbiology, 59(4). https://doi.org/10.1128/jcm.02402-20.
Safari, F., Afarid, M., Rastegari, B., Borhani-Haghighi, A., Barekati-Mowahed, M., &
Behzad-Behbahani, A. (2021). CRISPR systems: Novel approaches for detection and com-
bating COVID-19. Virus Research, 294, 198282. https://doi.org/10.1016/j.virusres.2020.
198282.
Sexton, N. R., Smith, E. C., Blanc, H., Vignuzzi, M., Peersen, O. B., & Denison, M. R. (2016).
Homology-based identification of a mutation in the coronavirus RNA-dependent RNA
polymerase that confers resistance to multiple mutagens. Journal of Virology, 90(16),
7415–7428. https://doi.org/10.1128/jvi.00080-16.
Shahbazi, R., Sghia-Hughes, G., Reid, J. L., Kubek, S., Haworth, K. G., Humbert, O., et al.
(2019). Targeted homology-directed repair in blood stem and progenitor cells with
CRISPR nanoformulations. Nature Materials, 18(10), 1124–1132. https://doi.org/
10.1038/s41563-019-0385-5.
Shmakov, S., Abudayyeh, O. O., Makarova, K. S., Wolf, Y. I., Gootenberg, J. S.,
Semenova, E., et al. (2015). Discovery and functional characterization of diverse class
150 CHAPTER 4 CRISPR use in diagnosis and therapy for COVID-19
1 Introduction
The coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syn-
drome coronavirus 2 (SARS-CoV-2) and is one of the most difficult health crises that
humanity has faced in recent years. The pandemic has affected millions of people
across the globe causing harm to humans as well as the economies of nations. Several
public health strategies such as the use of masks, social distancing, regular washing
of hands as well as contact tracing, have been employed since the beginning of the
outbreak to curtail the spread of the virus. However, these practices have not been
able to completely prevent the widespread of the pandemic (Young, Thone, & Jik,
2021). Despite the tireless efforts of researchers and scientists all over the world,
there is as of now, still, no cure for COVID-19, although the United States Food
and Drugs Administration (FDA) recently approved the use of remdesivir for
treatment, especially in severe cases of viral infection (Campos et al., 2020). The
outbreak of the pandemic has stretched the limits of healthcare systems and chal-
lenged the management of the situation using conventional tools in the development
Methods in Microbiology, Volume 50, ISSN 0580-9517, https://doi.org/10.1016/bs.mim.2022.03.001
Copyright © 2022 Elsevier Ltd. All rights reserved.
151
152 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
The S proteins have polymers that get embedded in the virion, obtaining the crown-like
appearance, which helps the virus connect to the host cell’s surface receptors and com-
bines the viral and host cell membranes, allowing the virus to enter the host cell
(Boopathi, Poma, & Kolandaivel, 2020). The spike protein (S-protein) facilitates
the coronavirus access into host cells (Wang, Hu, et al., 2020; Wang, Peng, et al.,
2020; Wang, Zhao, et al., 2020) by attaching to the ACE-2 receptor which has high
expression on some cell types, such as endothelial cells, alveolar cells, kidney cells,
intestinal epithelial cells, monocytes/macrophages, in addition to neuro-epithelial cells
and neurons (Wan et al., 2020; Zhou et al., 2020) and low-to-no expression, in most
cells and organs of the immune system (blood cells, spleen, bone marrow, and blood
vessels) (Garciá, Mancilla-Galindo, Paredes-Paredes, Tiburcio, & Ávila-Vanzzini,
2021). After the spike (S) protein binds to the ACE-2 receptor, cleavage by transmem-
brane proteases serine 2, cathepsin, or furin causes endocytosis and translocation of the
SARS-CoV-2 into endosomes (Reza-Zaldıvar, 2021), or direct viral envelope fusion
with host cell membrane for cell entrance (Sanclemente-Alaman et al., 2020). Since
the SARS-CoV-2 S protein is highly glycosylated and remains mostly in a closed pre-
fusion conformation (Turoňová et al., 2020), pre-activation of the S protein by furin
protease is thought to be an essential step to expose its receptor-binding domain
(Garciá et al., 2021).
Initial studies about COVID-I9 reported the effect of the disease mainly on the re-
spiratory tract but also on the gastrointestinal tract (Fig. 1). However, more recent
reports strongly suggest that the disease can lead to several extra-pulmonary compli-
cations and even multiple organ injuries. There have been reports of myocardial in-
flammation and venous thromboembolism especially with patients with pre-existing
cardiovascular conditions (Sarkesh et al., 2020). Hematological complications such
as lymphopenia, increased level of interleukin 6 (IL-6) and some cytokines, throm-
bocytopenia, neutrophilia caused by hyper-inflammatory and cytokine storm result-
ing in severe organ malfunction and physiologic decompensation have been reported
in patients with severe symptoms of COVID-19 (Alexandrova, Beykov, Vassilev,
Jukic, & Podlipnik, 2021). Viral infections have over the years been known to cause
serious structural and functional damage to the nervous system (Rahman et al., 2020;
Sarkesh et al., 2020). Several studies have reported some central nervous system
(CNS) complications such as cerebral infarctions in patients with severe cases of
COVID-19 disease (Macera, De Angelis, Sagnelli, & Coppola, 2020). Also, older pa-
tients particularly those with underlying ailments, are at high risk of impaired con-
sciousness or delirium at the onset of acute infections. These patients also display
symptoms like encephalopathy and confusion. Rare, nonspecific neurological symp-
toms have been reported in COVID-19 patients, including dizziness, headache (Fig. 1),
muscle injury leading to myalgia, neuralgia, and ataxia (Harrison, Lin, & Wang, 2020).
Several SARS COV2 infected patients have been reported to present with several
muscular disorders, some of which include myopathy, acute quadriplegic myopathy,
thick filament myopathy, and necrotizing myopathy (Gálvez-Barrón et al., 2021; Mba,
Sharndama, Osondu-chuka, & Okeke, 2021). Although the aged population is partic-
ularly vulnerable to COVID-19, due to their weakened immune system, poor ability to
heal damaged epithelium, and a reduced mucociliary clearance, which permits the
virus to easily move to the lung’s gas exchange components, children and young adults
can also be infected by the virus (Chao et al., 2020; Ho et al., 2001) (Fig. 1).
FIG. 2
Common vaccine strategies. This is a diagrammatic representation of the common vaccine
strategies been explored in the development of COVD-19 vaccines.
There are two major components of vaccines among others that should be given
great consideration during the production process. Firstly, protein or polysaccharide
antigens, which are supplied to or produced by the vaccine recipient. Another impor-
tant component is an infection signal that alerts and activates the host immune
system. Live attenuated vaccines can naturally provide both the antigen and the sig-
nal, while non-viral vaccine platforms can supply the antigens but frequently require
adjuvant that gives the danger signals to the immune system. Fig. 3 further illustrates
other basic components of a vaccine formulation.
Stabilizers
Active ingredients Stabilizers such as sugar or gelatin
Viral or bacterial antigens that are usually included to help
directly stimulate the immune maintain the efficacy of the vaccine
system with the primary aim of until ladministered.
preventing a full blown infection by
the causative organism.
Preservatives
Adjuvants Some vaccine formulations
These are diverse compounds contain preservatives (eg
added in very small quantities in thimerosal) which serves to
order to enhance the immune prevent contamination by
response to the vaccine dangerous microorganisms.
formulation.. VACCINE
Trace components
Residual inactivating ingredients
Antibiotics such as formaldehyde, and
Prevent contamination by bacteria residual cell culture materials
during the vaccine manufacturing (present in small quantities that
process. does not pose a safety concern).
Created in BioRender.com
FIG. 3
Basic constituents of vaccines.
158 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
virulence genes. The mutants lose their pathogenicity property in the host cells but
can replicate to a limited extent. Several genes possessed by coronaviruses are not
required for replication and these genes can be deleted, leading to attenuation in vivo.
Various non-structural proteins and structural E proteins can be deleted to yield vac-
cine strains of several coronaviruses. Deletion of the E protein leads to attenuation
and generation of an efficacious vaccine strain (Almazán et al., 2013; Netland et al.,
2010). There can be a problem of reversion of the attenuated strain to the virulent
strain (Jimenez-Guardeño et al., 2015). Therefore, the deletion of virulence genes
may provide a more effective mechanism of attenuation.
Codon deoptimization is another efficient approach to viral attenuation. The
nucleic acid sequence is modified to use suboptimal codon pairs to encode the
wild-type amino acid sequence, which considerably slows the translation of the viral
protein during infection. This approach can produce a replication-competent but
highly attenuated strain in vivo (Mueller et al., 2020). The recoded virus has an an-
tigenic property similar to that of its pathogenic parents. Consequently, the attenu-
ated viruses induce immune responses that are identical to those of virulent strains
(Mueller et al., 2020).
The inability of the attenuated strain to revert genetically to become pathogenic is
an important consideration in the generation of a live attenuated vaccine. Corona-
viruses are known to often recombine in nature and this makes the development
of an attenuated live vaccine against SARS-CoV-2 challenging. The attenuated
strain could recombine with other wild coronaviruses resulting in a fully virulent
strain. One of the drawbacks of live attenuated viruses is the use of exhaustively
long cell or animal cultures in their development. Also, pre-existing cross-reactive
immunity resulting from natural exposure with other human coronaviruses could
limit the efficacy of SARS-CoV-2 vaccines developed using this platform. Another
disadvantage of this technique is that attenuated vaccines cannot be given to
immune-compromised persons since the attenuated agent would find a niche to
multiply uncontrollably and, on rare occasions, revert to a wild-type phenotype,
resulting in severe disease. As a result of these drawbacks, only two live attenuated
SARS-CoV-2 vaccine candidates have reached clinical trials as seen in Table 1
(World Health Organization, 2021).
Continued
160 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
Continued
162 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
antigen. Inactivated viral vaccine also requires the administration of more than one
dose to be effective. There are sixteen inactivated SARS-CoV-2 vaccines under clin-
ical trials as seen in Table 1 (World Health Organization, 2021).
PS Protein subunit
DNA DNA
2%
16%
34% VVr Viral Vector (replicating)
1%
2%
5% VLP Virus Like Particle
9%
14% 2%
VVnr Viral Vector (non-replicating)
FIG. 4
Percentage representation of vaccine candidates in clinical development. Given is a statistical
representation of the different vaccine candidates currently under clinical development.
164 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
vector can reduce the magnitude of the triggered immune responses. In line with this,
administration of booster doses where it is needed becomes challenging as antibodies
against the viral vector produced after the prime vaccination can reduce the immu-
nological response. This problem can be circumvented by using a viral vector with
low human sero-prevalence such as ChAd (adenovirus derived from chimpanzees)
(Jeyanathan et al., 2020). Although this strategy for vaccine development is new, it
has been widely investigated for use in infectious diseases and cancer. The benefits
of this platform include safety and the ability to induce strong T-cell responses with-
out the need for an adjuvant (Draper & Heeney, 2010). Replicating viral vector vac-
cines need to be administered only once for protection and have a natural tropism
for the respiratory mucosa meaning that they are suited for respiratory tract vacci-
nation (Afkhami, Yao, & Xing, 2016). There is already existing technology for their
large-scale manufacturing and storage. Recombinant viral vectors are one of the
most common platforms for COVID-19 vaccine development, with eighteen candi-
dates currently in clinical trials (Fig. 4).
BioNTech COVID-19 vaccine is also based on this platform. One of the major draw-
backs of mRNA vaccines is the storage temperature of between 70 °C and 20 °C
which complicates the distribution logistics of these vaccines especially in develop-
ing countries. There are eighteen mRNA-based COVID-19 vaccines in clinical trials
(Fig. 4). Plasmid DNA vaccines share several characteristics with mRNA vaccines,
including safety, ease of production, and scalability (Hobernik & Bros, 2018). DNA
molecules are more stable than mRNA and can be stored at 4 °C, thereby simplifying
the storage and distribution of this type of vaccine. Some of the major limitations of
DNA vaccines are poor immunogenicity, repeated administration, and the need for
an adjuvant. Currently, there are ten DNA-based COVID-19 vaccines in clinical
trials as shown in Fig. 4, with some examples represented in Table 1.
1208
S2`
SS RBD SD2 HR1 CD TM
1 1273
NTD SD1 CH CT
HR2
S1/S2 FP
FIG. 5
Spike protein structure in SARS-CoV-2. SS, signal sequence; S20 , protease cleavage site; FP,
fusion peptide; HR1, heptad repeat 1; CH, central helix; CD, connector domain; HR2, heptad
repeat 2; TM, transmembrane domain; CT, cytoplasmic tail.
Adapted from Kheirandish et al., 2021.
NTD RBD FP CD
Membrane (M) protein
Membrane stabilization S1 S2
SARS-CoV-2
S1
Nucleocapsid (N) protein
RNA replication ACE2
mRNA replication
Host cell
Viral budding
FIG. 6
Diagrammatic representation of the SARS-CoV-2 structural proteins (Young et al., 2021).
Reproduced by permission of the Elsevier Permissions.
ACE2 receptor (Fig. 6), followed by the release of the viral pack in the host cell cy-
toplasm after the interaction with endosomal proteases. In fact, research has pinned
the high transmission rate of SARS-CoV-2 to the high affinity of its RBD to bind to
ACE2 receptors, an affinity that is 10–20 times higher than that of the SARS-CoV
virus. Cytokine storm which is a characteristic severe immune response in COVID-
19 disease is as a result of the endocytosis of SARS-CoV-2 into lung cells (Zarandi
et al., 2021).
Currently, most pre-clinical and clinical researches on vaccine development are
based on S protein. About 35 of the 47 vaccine candidates undergoing clinical trials
are based on S protein. Components of the S protein exploited as antigens include the
full-length S protein, the RBD domain, the S1, and S2 subunits, the N-terminal
domain (NTD), and the membrane fusion peptide (Ahmad et al., 2021). The infor-
mation acquired during SARS and MERS vaccine development in 2002 and 2012,
168 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
as well as binds the MHC II molecule using its CD4+ receptor (Shin et al., 2020).
These reactions cause T-helper cells to release interleukins which causes B cells
to proliferate. In addition to the proliferation of B cells, is its differentiation into
plasma cells, which then releases specific antibodies to the S protein fragment.
The interleukins, in turn, cause the initially produced T-helper cells to proliferate
and form T-helper memory cells. Both the T-helper cells and plasma cells produce
antibodies against some domains of the S protein of SARS-CoV-2 during future at-
tacks (De Soto, 2021). Furthermore, through the MHC I complex, cytotoxic T cells
are able to bind S protein fragments expressed by non-immune cells. This interaction
leads to the release of cytokines by cytotoxic cells and the proliferation and differ-
entiation of B cells into plasma cells. Additionally, the interaction of the S protein
with cytotoxic T cells paves the way for the elimination of cells infected by the
S protein of the SARS-CoV-2 virus in the future (De Soto, 2021).
the spike S1 subunit, membrane protein, and N protein is being developed by the
National Research Centre in Egypt, however, it is still undergoing pre-clinical trials
(Zarandi et al., 2021).
Ozarowski, Mrozikiewicz, & Wolski, 2021; Zarandi et al., 2021). Unlike other
structural proteins, the E protein is not a usual target for vaccines since they are not
sufficiently immunogenic because of its structure and low quantity (Karpinski
et al., 2021; Young et al., 2021). Nonetheless, a recent study by Abdelmageed and
co-researchers targeted a multiepitope-based peptide vaccine against the E protein
of human COVID-19 (Abdelmageed et al., 2020). There is a need, however, to thor-
oughly investigate the vaccine to determine its immunogenicity and safety profile
(Ahmad et al., 2021).
preparations the ability for easy targeted uptake in desired cells or tissues thus trigger-
ing an optimal immune response against the virus (Shin et al., 2020).
reduce the need for cold chain storage (O’Shea, Prausnitz, & Rouphael, 2021; Tran
et al., 2020). A study carried out by Kim et al. succeeded in formulating a dissolvable
microneedle matrix delivering SARS-CoV-2 antigen, and it was tested in a mice
model. Vaccine-induced antibodies were generated in significant amounts within
2 weeks post-administration (Kim et al., 2020). In another study, the RBD domain
peptide of the SARS-CoV-2 spike protein was delivered through a microneedle-
based formulation to mice. An ample amount of T-cell response, as well as antibody
generation, was also reported (Kuwentrai et al., 2021). More research is still needed
to determine procedures necessary for upscaling production and commercialization
of a microneedle-based formulation as this will be very beneficial in developing
countries of the world (Kumar & Kumar, 2021).
from respiratory viral infections (Hogan et al., 2001) which may produce longer-
lasting immunity against SARS-CoV-2 (Young et al., 2021). Despite this reality, sev-
eral researchers are continuously working to develop mucosal as well as other vaccines
that have alternative delivery routes to conventional ones. The mucosal route of admin-
istration offers the benefit of a lower risk of systemic adverse effects of the vaccine
formulation and a needle-free vaccination, making them more patient-friendly and also
eliminating the need for a skilled health professional (Strizova, Smetanova,
Bartunkova, & Milota, 2021). Some reports have postulated that since the nasal cavity
is a major entry point for the SARS_CoV 2, the nasal associated lymphoid tissues
(NALT), could present a promising target for COVID-19 vaccine delivery (Pandey
et al., 2021). The NALT, majorly consisting of dendritic cells, macrophages, and lym-
phoid follicles on the activation have been largely implicated in the clearance of viral
pathogens from the mucous layer (Pandey et al., 2021). A study exploring an intranasal
vaccine candidate for SARS-CoV reported the induction of Trm cells in the lungs,
which are vital for protection from viral infection (Baric et al., 2016). Another study
by An et al. (2020) showed that a single dose of adenovirus type 5 vectored vaccine
encoding the receptor-binding domain of SARS-COV 2 S-protein administered nasally
was able to induce both systemic and local immune responses against the SARS-CoV2
in mice. This was attributed primarily to the induction of mucosal IgA and serum neu-
tralizing antibodies (An et al., 2020). Hassan et al. (2020) also developed a chimpanzee
adenovirus-vectored vaccine which encodes a perfusing stabilized S-protein (Chad-
SAR-CoV-2-S), for delivery via the nasal route. This formulation was reported to in-
duce very high levels of neutralizing antibodies while enhancing systemic and mucosal
immunoglobulin A (IgA) and T-cell responses, thus preventing infection of SARS-
CoV-2 in the upper and lower respiratory tracts (Hassan et al., 2020). China is currently
testing a COVID-19 vaccine designed as an intranasal spray. This vaccine is composed
of weakened flu viruses (H1N1, H3N2, and B) combined with segments of the SARS-
CoV2 s protein, which mimics infection of respiratory viruses and can stimulate im-
mune responses. This candidate promises to provide the advantage of easy scale-up of
industrial production and distribution (Abdellatif et al., 2021).
Also, the oral route of drug delivery being the most preferred due to patient com-
pliance and ease of administration has been studied in the delivery of COVID-19
vaccines. An oral COVID-19 vaccine candidate (VXA-CoV2-1), developed by
VAXART, USA, is designed as an orally administered recombinant coated tablet.
It comprises an enteric-coated tablet containing an adenoviral vector, encoding
for the genes for the S and N-proteins of the SARS-COV2. On administration,
VXA-CoV2-1 triggers a mucosal immune response for defence against viruses caus-
ing respiratory infections as well high titres of neutralizing antibodies against the
SARS-CoV2. Also, the study revealed that there was no sign of mucosal damage
or weight loss in the hamsters used. Due to the fact that this vaccine candidate targets
the N-protein in addition to the S-protein, it promises to provide better protection
against the new viral variants. This has recently received approval for phase II clin-
ical trials (Ashraf et al., 2021). A UK-based company, IsoBio, is currently develop-
ing the Oral Pro-COVID-19®. This candidate is a non-replicating viral vector
178 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
vaccine that expresses the S-protein, designed as a thermally stable capsulated form.
This is particularly important as it provides the advantage of self-administration and
needle-free application. In addition, the need for cold-chain storage is eliminated as
the vaccine is designed as a thermally stable capsule. This makes this candidate
especially interesting for developing countries with erratic power supply (Ashraf
et al., 2021).
delivery vehicles have been modified to stabilize the vaccine antigens being deliv-
ered (Wang, Hu, et al., 2020; Wang, Peng, et al., 2020; Wang, Zhao, et al., 2020).
Despite the benefits that nanotechnology offers in vaccine development, there are
also some issues of concern. One of the major problems of nano-based systems is the
issue of toxicity. There are several molecular mechanisms of toxicity elicited by
nanosystems. Some of these systems have been known to interact with cellular
DNA, interrupting important enzyme functions and thus causing harm to the organ-
ism. There have also been some reports of these systems generating reactive oxygen
species that eventually cause harm to genetic materials or disrupt vital enzyme func-
tions (Pandey et al., 2021). Also, because the process of vaccine development testing
and regulation is long, taking about 10–15 years, and the current COVID-19 vaccine
candidates have been developed within a space of a year, there are concerns about the
safety and long term effects of these formulations (Soleimanpour & Yaghoubi,
2021). The use of a multi-component nano-based vaccine, having complex structural
make-up (Bonam et al., 2021) can bring about an increased cost of production as a
result of a rigorous process of production. Some reports have also argued that since
many nano-vaccines are usually produced in small batches for research, the scale-up
of these systems might be challenging. This is because this process is largely plagued
by variations in size, shapes as well as other properties (Kim et al., 2014). It is how-
ever advocated that self-assemble nano-based vaccine systems be developed to
tackle the many challenges of large-scale production protocols. Also, the process
of surface modification of nano-carriers for vaccines usually involves a time-
consuming, costly and complicated process of purification (Kim et al., 2014).
are based on S protein. This study has provided updates on the current and advanced
nanotechnological strategies employed for the development of COVID-19 vaccines.
The nucleic acid vaccine candidate (mRNA-1273) by Moderna and currently in use
is encapsulated in lipid nanoparticles carriers. This typifies the use of nano-based
systems as effective antigen delivery systems. This review also explored several
nano-based adjuvants and novel delivery devices employed in the development of
COVID-19 vaccines. Also, the application of nanotechnology in the development
of these vaccine candidates has created a wide range of formulations that could
be delivered via alternative routes of administrations to the commonly used intra-
muscular route. The use of nanotechnological strategies in the development of
COVID-19 vaccines is still at the incipient stage as very few of these systems are
commercially available. However, these nano-based systems are relevant tools that
could play an excellent role in combating the pandemic.
References
Abdellatif, A. A. H., Tawfeek, H. M., Abdelfattah, A., El-Saber Batiha, G., & Hetta, H. F.
(2021). Recent updates in COVID-19 with emphasis on inhalation therapeutics: Nano-
structured and targeting systems. Journal of Drug Delivery Science and Technology,
63, 102435. https://doi.org/10.1016/j.jddst.2021.102435.
Abdelmageed, M. I., Abdelmoneim, A. H., Mustafa, M. I., Elfadol, N. M., Murshed, N. S.,
Shantier, S. W., et al. (2020). Design of a Multiepitope-Based Peptide Vaccine against
the e protein of human COVID-19: An Immunoinformatics approach. BioMed Research
International, 2020. https://doi.org/10.1155/2020/2683286.
Afkhami, S., Yao, Y., & Xing, Z. (2016). Methods and clinical development of adenovirus-
vectored vaccines against mucosal pathogens. Molecular Therapy - Methods and Clinical
Development, 3, 16030. https://doi.org/10.1038/mtm.2016.30.
Ahmad, J., Hanif, A., Farooqi, T., Hyder, F., Anwar, S., & Rengasamy, K. R. R. (2021). Bio-
medicine & Pharmacotherapy Targets and strategies for vaccine development against
SARS-CoV-2. Biomedicine & Pharmacotherapy, 137, 111254. https://doi.org/10.1016/
j.biopha.2021.111254.
Ahmed, E., Sajjad, N., Ali, A., Aldakeel, F. M., Mateen, A., Alqahtani, M. S., et al. (2021).
International Immunopharmacology SARS-CoV-2 : Insight in genome structure, patho-
genesis and viral receptor binding analysis – An updated review. International Immuno-
pharmacology, 95, 107493. https://doi.org/10.1016/j.intimp.2021.107493.
Alexandrova, R., Beykov, P., Vassilev, D., Jukic, M., & Podlipnik, C. (2021). The virus that
shook the world: Questions and answers about SARS-CoV-2 and COVID-19. Biotechnol-
ogy and Biotechnological Equipment, 35(1), 74–102. https://doi.org/10.1080/
13102818.2020.1847683.
Alimardani, V., Abolmaali, S. S., & Tamaddon, A. M. (2021). Recent advances on
nanotechnology-based strategies for prevention, diagnosis, and treatment of coronavirus
infections. Journal of Nanomaterials, 2021. https://doi.org/10.1155/2021/9495126.
Almazán, F., Dediego, M. L., Sola, I., Zuñiga, S., Nieto-Torres, J. L., Marquez-Jurado, S., et al.
(2013). Engineering a replication-competent, propagation-defective middle east respira-
tory syndrome coronavirus as a vaccine candidate. MBio, 4(5), 1–11. https://doi.org/
10.1128/mBio.00650-13.
References 181
Chao, J. Y., Derespina, K. R., Herold, B. C., Goldman, D. L., Aldrich, M., Weingarten, J., et al.
(2020). Clinical characteristics and outcomes of hospitalized and critically ill children and
adolescents with coronavirus disease 2019 at a tertiary care medical Center in new York
City. Journal of Pediatrics, 223(August), 14–19.e2. https://doi.org/10.1016/j.jpeds.
2020.05.006.
Chauhan, G., Madou, M. J., Kalra, S., Chopra, V., Ghosh, D., & Martinez-Chapa, S. O. (2020).
Nanotechnology for COVID-19: Therapeutics and vaccine research. ACS Nano, 14(7),
7760–7782. https://doi.org/10.1021/acsnano.0c04006.
Chen, Y., Liu, Q., & Guo, D. (2020). Emerging coronaviruses: Genome structure, replication,
and pathogenesis. Journal of Medical Virology, 92(4), 418–423.
Christensen, D., Smith, K., Andersen, P., & Agger, E. M. (2011). Cationic liposomes as vac-
cine adjuvants. Expert Review of Vaccines, 10(4), 513–521.
Chung, Y. H., Beiss, V., Fiering, S. N., & Steinmetz, N. F. (2020). Covid-19 vaccine frontrun-
ners and their nanotechnology design. ACS Nano, 14(10), 12522–12537. https://doi.org/
10.1021/acsnano.0c07197.
Donato, S. D., Grano, D., Minutello, M., Lecchi, G., Faccini, M., & Agnello, M. (1999). Safety
and immunogenicity of MF59-adjuvanted influenza vaccine in the elderly. Vaccine, 17,
3094–3101.
De Soto, J. A. (2021). Evaluation of the Moderna, Pfizer/biotech, Astrazeneca/Oxford and
sputnik V vaccines for Covid-19. Advance Research Journal of Medical Clinical and Sci-
ence, 07(01), 408–414. https://doi.org/10.15520/arjmcs.v7i01.246.
DeDiego, M. L., Álvarez, E., Almazán, F., Rejas, M. T., Lamirande, E., Roberts, A., et al.
(2007). A severe acute respiratory syndrome coronavirus that lacks the E gene is attenu-
ated in vitro and in vivo. Journal of Virology, 81(4), 1701–1713. https://doi.org/10.1128/
jvi.01467-06.
Draper, S. J., & Heeney, J. L. (2010). Viruses as vaccine vectors for infectious diseases and
cancer. Nature Reviews Microbiology, 8(1), 62–73. https://doi.org/10.1038/nrmicro2240.
Erasmus, J. H., Med, S. T., Erasmus, J. H., Khandhar, A. P., Connor, M. A. O., Walls, A. C.,
et al. (2020). An alphavirus-derived replicon RNA vaccine induces SARS-CoV-2 neutral-
izing antibody and T cell responses in mice and nonhuman primates. Science Translational
Medicine, 12(555), 1–17. https://doi.org/10.1126/scitranslmed.abc9396.
Gálvez-Barrón, C., Arroyo-Huidobro, M., Miňarro, A., Añaños, G., Chamero, A., Martı́n, M.,
et al. (2021). COVID-19: Clinical presentation and prognostic factors of severe disease
and mortality in the oldest-old population: A cohort study. Gerontology, 1–14. https://
doi.org/10.1159/000515159.
Garciá, M. M. A., Mancilla-Galindo, J., Paredes-Paredes, M., Tiburcio, Á. Z., & Ávila-
Vanzzini, N. (2021). Mechanisms of infection by SARS-CoV-2, inflammation and poten-
tial links with the microbiome. Future Virology, 16(1), 43–57. https://doi.org/10.2217/fvl-
2020-0310.
Ghaffari, M., Mollazadeh-Bajestani, M., Moztarzadeh, F., Uluda g, H., Hardy, J. G., &
Mozafari, M. (2021). An overview of the use of biomaterials, nanotechnology, and stem
cells for detection and treatment of COVID-19: Towards a framework to address future
global pandemics. Emergent Materials, 4(1), 19–34. https://doi.org/10.1007/s42247-
020-00143-9.
Grifoni, A., Weiskopf, D., Ramirez, S. I., Mateus, J., Dan, J. M., Moderbacher, C. R., et al.
(2020). Targets of T cell responses to SARS-CoV-2 coronavirus in humans with
COVID-19 disease and unexposed individuals. Cell. https://doi.org/10.1016/j.cell.2020.
05.015.
References 183
Hagan, D. T. O. (2007). MF59 is a safe and potent vaccine adjuvant that enhances protection
against influenza virus infection. Expert Review of Vaccines, 6(5), 699–710. https://doi.
org/10.1586/14760584.6.5.699.
Harrison, A. G., Lin, T., & Wang, P. (2020). Mechanisms of SARS-CoV-2 transmission and
pathogenesis. Trends in Immunology, 41(12), 1100–1115. https://doi.org/10.1016/j.
it.2020.10.004.
Hassan, A. O., Kafai, N. M., Dmitriev, I. P., Fox, J. M., Smith, B. K., Harvey, I. B., et al.
(2020). A single-dose intranasal ChAd vaccine protects upper and lower respiratory tracts
against SARS-CoV-2. Cell, 183(1), 169–184. https://doi.org/10.1016/j.cell.2020.08.026.
He, Y., Zhou, Y., Wu, H., Luo, B., Chen, J., Li, W., et al. (2004). Identification of Immuno-
dominant sites on the spike protein of severe acute respiratory syndrome (SARS) corona-
virus: Implication for developing SARS diagnostics and vaccines. The Journal of
Immunology, 173(6), 4050–4057. https://doi.org/10.4049/jimmunol.173.6.4050.
Ho, J. C., Hu, W. H. C., Lam, W. K., Chu, L. W., Zheng, L., Tipoe, G. L., et al. (2001). The
effect of aging on nasal mucociliary clearance, beat frequency and ultrastructure of respi-
ratory cilia. In The 6th Medical Research Conference, Hong Kong, China, 23(13–14)
(p. 66).
Hobernik, D., & Bros, M. (2018). DNA vaccines—How far from clinical use? International
Journal of Molecular Sciences, 19(11), 1–28. https://doi.org/10.3390/ijms19113605.
Hogan, R. J., Usherwood, E. J., Zhong, W., Roberts, A. A., Dutton, R. W., Harmsen, A. G.,
et al. (2001). Activated antigen-specific CD8 + T cells persist in the lungs following recov-
ery from respiratory virus infections. Journal of Immunology, 166, 1813–1822.
Hurst, K. R., Koetzner, C. A., & Masters, P. S. (2009). Identification of in vivo-interacting
domains of the murine coronavirus Nucleocapsid protein. Journal of Virology, 83(14),
7221–7234. https://doi.org/10.1128/jvi.00440-09.
Jackson, L. A., Anderson, E. J., Rouphael, N. G., Roberts, P. C., Makhene, M., Coler, R. N., et al.
(2020). An mRNA vaccine against SARS-CoV-2 — Preliminary report. New England Jour-
nal of Medicine, 383(20), 1920–1931. https://doi.org/10.1056/NEJMoa2022483.
Jackson, N. A. C., Kester, K. E., Casimiro, D., Gurunathan, S., & DeRosa, F. (2020). The
promise of mRNA vaccines: A biotech and industrial perspective. Npj Vaccines, 5(1),
3–8. https://doi.org/10.1038/s41541-020-0159-8.
Jeyanathan, M., Afkhami, S., Smaill, F., Miller, M. S., Lichty, B. D., & Xing, Z. (2020). -
mmunological considerations for COVID-19 vaccine strategies. Nature Reviews Immunol-
ogy, 20(October), 615–632. https://doi.org/10.1038/s41577-020-00434-6.
Jiang, S., Hillyer, C., & Du, L. (2020). Jiang S, Hillyer C, Du L. neutralizing antibodies against
SARS- CoV-2 and other human coronaviruses. Trends in Immunology, 41(5), 355–359.
https://doi.org/10.1016/j.it.2020.03.007.
Jimenez-Guardeño, J. M., Regla-Nava, J. A., Nieto-Torres, J. L., DeDiego, M. L., Castaño-
Rodriguez, C., Fernandez-Delgado, R., et al. (2015). Identification of the mechanisms
causing reversion to virulence in an attenuated SARS-CoV for the Design of a Genetically
Stable Vaccine. PLoS Pathogens, 11(10), 1–36. https://doi.org/10.1371/journal.
ppat.1005215.
Karpinski, T. M., Ozarowski, M., Mrozikiewicz, A. S., & Wolski, H. (2021). Theranostics the
2020 race towards SARS-CoV-2 specific vaccines. Theranostics, 11(4), 1690–1702.
https://doi.org/10.7150/thno.53691.
Keech, C., Albert, G., Cho, I., Robertson, A., Reed, P., Neal, S., et al. (2020). Phase 1–2 trial of
a SARS-CoV-2 recombinant spike protein nanoparticle vaccine. New England Journal of
Medicine, 383(24), 2320–2332. https://doi.org/10.1056/nejmoa2026920.
184 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
Kim, E., Erdos, G., Huang, S., Kenniston, T., Balmert, S., Carey, C., et al. (2020). Microneedle
array delivered recombinant coronavirus vaccines: Immunogenicity and rapid transla-
tional development. eBioMedicine, 55, 102743.
Kim, M. G., Park, J. Y., Shon, Y., Kim, G., Shim, G., & Oh, Y. K. (2014). Nanotechnology and
vaccine development. Asian Journal of Pharmaceutical Sciences, 9(5), 227–235. https://
doi.org/10.1016/j.ajps.2014.06.002.
Ksiazek, T. G., Erdman, D., Goldsmith, C. S., Zaki, S. R., Peret, T., Emery, S., et al. (2003).
A novel coronavirus associated with severe acute respiratory syndrome. New England
Journal of Medicine, 348(20), 1953–1966. https://doi.org/10.1056/NEJMoa030781.
Kumar, A., & Kumar, A. (2021). Mucosal and transdermal vaccine delivery strategies against
COVID-19. Drug Delivery and Translational Research, 0123456789. https://doi.org/
10.1007/s13346-021-01001-9.
Kusi, K. A., Remarque, E. J., Riasat, V., Walraven, V., Thomas, A. W., Faber, B. W., et al.
(2011). Safety and immunogenicity of multi-antigen AMA1-based vaccines formulated
with CoVaccine HT™ and Montanide ISA 51 in rhesus macaques. Malaria Journal,
10(1), 182. https://doi.org/10.1186/1475-2875-10-182.
Kuwentrai, C., Yu, J., Rong, L., Zhang, B. Z., Hu, Y. F., Gong, H. R., et al. (2021). Intradermal
delivery of receptor-binding domain of SARS-CoV-2 spike protein with dissolvable
microneedles to induce humoral and cellular responses in mice. Bioengineering and
Translational Medicine, 6(1), e10202. https://doi.org/10.1002/btm2.10202.
Lehrer, A. T., Wong, T. A. S., Lieberman, M. M., Humphreys, T., Clements, D. E.,
Bakken, R. R., et al. (2018). Recombinant proteins of Zaire ebolavirus induce potent
humoral and cellular immune responses and protect against live virus infection in mice.
Vaccine, 36(22), 3090–3100. https://doi.org/10.1016/j.vaccine.2017.01.068.
Lokugamage, K. G., Yoshikawa-Iwata, N., Ito, N., Watts, D. M., Wyde, P. R., Wang, N., et al.
(2008). Chimeric coronavirus-like particles carrying severe acute respiratory syndrome
coronavirus (SCoV) S protein protect mice against challenge with SCoV. Vaccine,
26(6), 797–808. https://doi.org/10.1016/j.vaccine.2007.11.092.
Lutz, J., Lazzaro, S., Habbeddine, M., Schmidt, K. E., Baumhof, P., Mui, B. L., et al. (2017).
Unmodified mRNA in LNPs constitutes a competitive technology for prophylactic
vaccines. Npj Vaccines, 2(1), 1–9. https://doi.org/10.1038/s41541-017-0032-6.
Macera, M., De Angelis, G., Sagnelli, C., & Coppola, N. (2020). Clinical presentation of
covid-19: Case series and review of the literature. International Journal of Environmental
Research and Public Health, 17(14), 1–11. https://doi.org/10.3390/ijerph17145062.
Machhi, J., Shahjin, F., Das, S., Patel, M., Abdelmoaty, M. M., Cohen, J. D., et al. (2021).
Nanocarrier vaccines for SARS-CoV-2. Advanced Drug Delivery Reviews, 171(January),
215–239. https://doi.org/10.1016/j.addr.2021.01.002.
Magnusson, S. E., Altenburg, A. F., Bengtsson, K. L., Bosman, F., de Vries, R. D.,
Rimmelzwaan, G. F., et al. (2018). Matrix-M™ adjuvant enhances immunogenicity of
both protein- and modified vaccinia virus Ankara-based influenza vaccines in mice.
Immunologic Research, 66(2), 224–233. https://doi.org/10.1007/s12026-018-8991-x.
Malabadi, R. B., Meti, N. T., & Chalannavar, R. K. (2021). Applications of nanotechnology in
vaccine development for coronavirus (SARS-CoV-2) disease (Covid-19). International
Journal of Research and Scientific Innovation (IJRSI), 8(2), 191–198. Retrieved from
www.rsisinternational.org.
Mason, R. J. (2020). Pathogenesis of COVID-19 from a cell biology perspective. European
Respiratory Journal. https://doi.org/10.1183/13993003.00607-2020.
References 185
Mba, I. E., Sharndama, H. C., Osondu-chuka, G. O., & Okeke, O. P. (2021). Immunobiology
and nanotherapeutics of severe acute respiratory syndrome 2 (SARS-CoV-2): A current
update. Infectious Diseases, 53(8), 559–580. https://doi.org/10.1080/23744235.2021.
1916071.
McKay, P. F., Hu, K., Blakney, A. K., Samnuan, K., Bouton, C. R., Rogers, P., et al. (2020).
Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine induces equivalent preclin-
ical antibody titers and viral neutralization to recovered COVID-19 patients. BioRxiv,
1–14. https://doi.org/10.1101/2020.04.22.055608.
Menon, I., Bagwe, P., Gomes, K. B., Bajaj, L., Gala, R., Uddin, M. N., et al. (2021). Micro-
needles: A new generation vaccine delivery system. Micromachines, 12(4), 1–18. https://
doi.org/10.3390/mi12040435.
Mueller, S., Stauft, C. B., Kalkeri, R., Koidei, F., Kushnir, A., Tasker, S., et al. (2020).
A codon-pair deoptimized live-attenuated vaccine against respiratory syncytial virus is im-
munogenic and efficacious in non-human primates. Vaccine, xxxx, 2–7. https://doi.org/
10.1016/j.vaccine.2020.02.056.
Murdin, A. D., Barreto, L., & Plotkin, S. (1996). Inactivated poliovirus vaccine: Past and present
experience. Vaccine, 14(8), 735–746. https://doi.org/10.1016/0264-410X(95)00211-I.
Naskalska, A., Dabrowska, A., Nowak, P., Szczepanski, A., Jasik, K., Milewska, A., et al.
(2018). Novel coronavirus-like particles targeting cells lining the respiratory tract. PLoS
One, 13(9), 1–21. https://doi.org/10.1371/journal.pone.0203489.
Netland, J., DeDiego, M. L., Zhao, J., Fett, C., Álvarez, E., Nieto-Torres, J. L., et al. (2010).
Immunization with an attenuated severe acute respiratory syndrome coronavirus deleted in
E protein protects against lethal respiratory disease. Virology, 399(1), 120–128. https://doi.
org/10.1016/j.virol.2010.01.004.
Nieto-Torres, J. L., DeDiego, M. L., Verdiá-Báguena, C., Jimenez-Guardeño, J. M.,
Regla-Nava, J. A., Fernandez-Delgado, R., et al. (2014). Severe acute respiratory
syndrome coronavirus envelope protein Ion Channel activity promotes virus fitness and
pathogenesis. PLoS Pathogens, 10(5), e1004077. https://doi.org/10.1371/journal.
ppat.1004077.
O’Shea, J., Prausnitz, M. R., & Rouphael, N. (2021). Dissolvable microneedle patches to en-
able increased access to vaccines against SARS-CoV-2 and future pandemic outbreaks.
Vaccine, 9(4), 320. https://doi.org/10.3390/vaccines9040320.
Pandey, A., Nikam, A. N., Mutalik, S. P., Fernandes, G., Shreya, A. B., Padya, B. S., et al.
(2021). Architectured therapeutic and diagnostic Nanoplatforms for combating SARS-
CoV-2: Role of inorganic, organic, and radioactive materials. ACS Biomaterials Science &
Engineering, 7(1), 31–54. https://doi.org/10.1021/acsbiomaterials.0c01243.
Pardi, N., Hogan, M. J., Porter, F. W., & Weissman, D. (2018). mRNA vaccines-a new era in
vaccinology. Nature Reviews Drug Discovery, 17(4), 261–279. https://doi.org/10.1038/
nrd.2017.243.
Petkar, K. C., Patil, S. M., Chavhan, S. S., Kaneko, K., Sawant, K. K., Kunda, N. K., et al.
(2021). An overview of nanocarrier-based adjuvants for vaccine delivery. Pharmaceutics,
13(4), 1–29. https://doi.org/10.3390/pharmaceutics13040455.
Petsch, B., Schnee, M., Vogel, A. B., Lange, E., Hoffmann, B., Voss, D., et al. (2012). Pro-
tective efficacy of in vitro synthesized, specific mRNA vaccines against influenza a virus
infection. Nature Biotechnology, 30(12), 1210–1216. https://doi.org/10.1038/nbt.2436.
Plotkin, S. A., Orenstein,, W. A., Offit, P. A., & Edwards, K. M. (2017). Vaccines E-book.
Elsevier Health Sciences.
186 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
Polack, F., Thomas, S., Kitchin, N., Absalon, J., Gurtman, A., Lockhart, S., et al. (2020).
Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. The New England Jour-
nal of Medicine, 10, 1–13. https://doi.org/10.1056/NEJMoa2034577.
Poon, C., Gallo, J., Joo, J., Chang, T., Bañobre-López, M., & Chung, E. J. (2018). Hybrid,
metal oxide-peptide amphiphile micelles for molecular magnetic resonance imaging of
atherosclerosis. Journal of Nanobiotechnology, 16(1), 1–11. https://doi.org/10.1186/
s12951-018-0420-8.
Qian, Z., Travanty, E. A., Oko, L., Edeen, K., Berglund, A., Wang, J., et al. (2012). Innate
immune response of human alveolar type II cells infected with severe acute respiratory
syndrome – Coronavirus. American Journal of Respiratory Cell and Molecular Biology,
10(22), 22–26. https://doi.org/10.1165/rcmb.2012-0339OC.
Rahman, H. S., Aziz, M. S., Hussein, R. H., Othman, H. H., Salih Omer, S. H., Khalid, E. S.,
et al. (2020). The transmission modes and sources of COVID-19: A systematic review.
International Journal of Surgery Open, 26, 125–136. https://doi.org/10.1016/j.
ijso.2020.08.017.
Rao, L., Xia, S., Xu, W., Tian, R., Yu, G., Gu, C., et al. (2020). Decoy nanoparticles protect
against COVID-19 by concurrently adsorbing viruses and inflammatory cytokines. Pro-
ceedings of the National Academy of Sciences of the United States of America,
117(44), 27141–27147. https://doi.org/10.1073/pnas.2014352117.
Raoufi, E., Bahramimeimandi, B., Salehi-Shadkami, M., Chaosri, P., & Mozafari, M. R.
(2021). Methodical design of viral vaccines based on avant-Garde nanocarriers:
A multi-domain narrative review. Biomedicine, 9(5), 1–20. https://doi.org/10.3390/
biomedicines9050520.
Reza-Zaldıvar, E. E., et al. (2021). Infection mechanism of SARS-COV-2 and its implication
on the nervous system. Frontiers in Immunology, 11, 621735. In press. https://doi.org/
10.3389/fimmu.2020.621735.
Ruiz-Hitzky, E., Darder, M., Wicklein, B., Ruiz-Garcia, C., Martı́n-Sampedro, R., del
Real, G., et al. (2020). Nanotechnology responses to COVID-19. Advanced Healthcare
Materials, 9(19), 1–26. https://doi.org/10.1002/adhm.202000979.
Sanclemente-Alaman, I., Moreno-Jimenez, L., Benito-Martı́n, M. S., Canales-Aguirre, A.,
Matı́as-Guiu, J. A., Matı́as-Guiu, J., et al. (2020). Experimental models for the study of
central nervous system infection by SARS-CoV-2. Frontiers in Immunology, 11(8),
1–12. https://doi.org/10.3389/fimmu.2020.02163.
Sarkesh, A., Sorkhabi, A. D., Sheykhsaran, E., Alinezhad, F., Mohammadzadeh, N.,
Hemmat, N., et al. (2020). Extrapulmonary clinical manifestations in COVID-19 patients.
American Journal of Tropical Medicine and Hygiene, 103(5), 1783–1796. https://doi.org/
10.4269/ajtmh.20-0986.
Schnee, M., Vogel, A. B., Voss, D., Petsch, B., Baumhof, P., Kramps, T., et al. (2016). An
mRNA vaccine encoding rabies virus glycoprotein induces protection against lethal infec-
tion in mice and correlates of protection in adult and newborn pigs. PLoS Neglected Trop-
ical Diseases, 10(6), 1–20. https://doi.org/10.1371/journal.pntd.0004746.
Shin, M. D., Shukla, S., Chung, Y. H., Beiss, V., Chan, S. K., Ortega-Rivera, O. A., et al.
(2020). COVID-19 vaccine development and a potential nanomaterial path forward.
Nature Nanotechnology, 15(8), 646–655. https://doi.org/10.1038/s41565-020-0737-y.
Soleimanpour, S., & Yaghoubi, A. (2021). COVID-19 vaccine: Where are we now and where
should we go?. Expert Review of Vaccines: Taylor and Francis Ltd. https://doi.org/
10.1080/14760584.2021.1875824.
References 187
Strizova, Z., Smetanova, J., Bartunkova, J., & Milota, T. (2021). Principles and challenges in
anti-COVID-19 vaccine development. International Archives of Allergy and Immunology,
182(4), 339–349. https://doi.org/10.1159/000514225.
Suganya, S., Divya, S., & Parani, M. (2021). Severe acute respiratory syndrome-coronavirus-2:
Current advances in therapeutic targets and drug development. Reviews in Medical
Virology. https://doi.org/10.1002/rmv.2174.
Tai, W., He, L., Zhang, X., Pu, J., Voronin, D., Jiang, S., et al. (2020). Characterization of the
receptor-binding domain (RBD) of 2019 novel coronavirus: Implication for development
of RBD protein as a viral attachment inhibitor and vaccine. Cellular and molecular immu-
nology, 17(6), 613–620. https://doi.org/10.1038/s41423-020-0400-4.
Tran, K. T. M., Gavitt, T. D., Farrell, N. J., Curry, E. J., Mara, A. B., Patel, A., et al. (2020).
Transdermal microneedles for the programmable burst release of multiple vaccine pay-
loads. Nature Biomedical Engineering. https://doi.org/10.1038/s41551-020-00650-4.
Turoňová, B., Sikora, M., Sch€urmann, C., Hagen, W. J. H., Welsch, S., Blanc, F. E. C., et al.
(2020). In situ structural analysis of SARS-CoV-2 spike reveals flexibility mediated by
three hinges. Science, 370(6513), 203–208. https://doi.org/10.1126/science.abd5223.
van Doremalen, N., Lambe, T., Spencer, A., Belij-Rammerstorfer, S., Purushotham, J. N.,
Port, J. R., et al. (2020). ChAdOx1 nCoV-19 vaccine prevents SARS-CoV-2 pneumonia
in rhesus macaques. Nature, 586(7830), 578–582. https://doi.org/10.1038/s41586-020-
2608-y.
Vellozzi, C., Burwen, D. R., Dobardzic, A., Ball, R., Walton, K., & Haber, P. (2009). Safety of
trivalent inactivated influenza vaccines in adults: Background for pandemic influenza vac-
cine safety monitoring. Vaccine, 27(15), 2114–2120. https://doi.org/10.1016/j.vaccine.
2009.01.125.
Wan, Y., Shang, J., Graham, R., Baric, R. S., & Li, F. (2020). Receptor recognition by the
novel coronavirus from Wuhan: An analysis based on decade-long structural studies of
SARS coronavirus. Journal of Virology, 94(7). https://doi.org/10.1128/jvi.00127-20.
Wang, D., Hu, B., Hu, C., Zhu, F., Liu, X., Zhang, J., et al. (2020). Clinical characteristics of
138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan,
China. JAMA : The Journal of the American Medical Association, 323(11), 1061–1069.
https://doi.org/10.1001/jama.2020.1585.
Wang, J., Peng, Y., Xu, H., Cui, Z., & Williams, R. O. (2020). The COVID-19 vaccine race:
Challenges and opportunities in vaccine formulation. AAPS PharmSciTech, 21(6), 225.
https://doi.org/10.1208/s12249-020-01744-7.
Wang, M. Y., Zhao, R., Gao, L. J., Gao, X. F., Wang, D. P., & Cao, J. M. (2020). SARS-CoV-2:
Structure, biology, and structure-based therapeutics development. Frontiers in Cellular
and Infection Microbiology. https://doi.org/10.3389/fcimb.2020.587269.
WHO. (2020). Draft landscape of COVID-19 candidate vaccines- November 3. World Heal.
Organ.
World Health Organization. (2021). Status of COVID-19 vaccines within WHO EUL/PQ eval-
uation process (20 January 2021). World Health Organization. (February), 2. Retrieved
from https://extranet.who.int/pqweb/sites/default/files/documents/Status_COVID_VAX_
20Jan2021_v2.pdf.
Wu, Z., & McGoogan, J. M. (2020). Characteristics of and important lessons from the coro-
navirus disease 2019 (COVID-19) outbreak in China: Summary of a report of 72314 cases
from the Chinese Center for Disease Control and Prevention. JAMA : The Journal of
the American Medical Association, 323(13), 1239–1242. https://doi.org/10.1001/
jama.2020.2648.
188 CHAPTER 5 Recent nano-technological strategies for COVID-19 vaccines
Wu, F., Wang, A., Liu, M., Wang, Q., Chen, J., Xia, S., et al. (2020). Neutralizing antibody
responses to SARS-CoV-2 in a COVID-19 recovered patient cohort and their implications.
SSRN Electronic Journal. https://doi.org/10.2139/ssrn.3566211.
Xia, S., Zhu, Y., Liu, M., Lan, Q., Xu, W., Wu, Y., et al. (2020). Fusion mechanism of 2019-
nCoV and fusion inhibitors targeting HR1 domain in spike protein. Cellular and molecular
immunology. https://doi.org/10.1038/s41423-020-0374-2.
Yang, D. (2021). Application of nanotechnology in the COVID-19 pandemic. International
Journal of Nanomedicine, 16, 623–649. https://doi.org/10.2147/IJN.S296383.
Young, J., Thone, M. N., & Jik, Y. (2021). COVID-19 vaccines : The status and perspectives in
delivery points of view. Advanced Drug Delivery Reviews, 170, 1–25. https://doi.org/
10.1016/j.addr.2020.12.011.
Zarandi, P. K., Zinatizadeh, M. R., Zinatizadeh, M., Yousefi, M. H., & Rezaei, N. (2021).
SARS-CoV-2: From the pathogenesis to potential anti-viral treatments. Biomedicine &
Pharmacotherapy, 137, 111352. https://doi.org/10.1016/j.biopha.2021.111352.
Zhang, N. N., Li, X. F., Deng, Y. Q., Zhao, H., Huang, Y. J., Yang, G., et al. (2020).
A thermostable mRNA vaccine against COVID-19. Cell, 182(5), 1271–1283. https://
doi.org/10.1016/j.cell.2020.07.024.
Zhang, D. Y., Zheng, Y., Tan, C. P., Sun, J. H., Zhang, W., Ji, L. N., et al. (2017). Graphene
oxide decorated with Ru(II)-polyethylene glycol complex for lysosome-targeted imaging
and photodynamic/Photothermal therapy. ACS Applied Materials and Interfaces, 9(8),
6761–6771. https://doi.org/10.1021/acsami.6b13808.
Zhao, L., Seth, A., Wibowo, N., Zhao, C.-X., Mitter, N., Yu, C., et al. (2014). Nanoparticle
vaccines. Vaccine, 32(3), 327–337. https://doi.org/10.1016/j.vaccine.2013.11.069.
Zhao, J., Yuan, Q., Wang, H., Liu, W., Liao, X., Su, Y., et al. (2020). Antibody responses to
SARS-CoV-2 in patients with novel coronavirus disease 2019. Clinical Infectious Dis-
eases, 71(16), 2027–2034. https://doi.org/10.1093/cid/ciaa344.
Zhou, P., Yang, X.-L., Wang, X.-G., Hu, B., Zhang, L., Zhang, W., et al. (2020). A pneumonia
outbreak associated with a new coronavirus of probable bat origin. Nature, 579(7798),
270–273. https://doi.org/10.1038/s41586-020-2012-7.
CHAPTER
A review of
hypersensitivity methods
to detect immune
responses to SARS-CoV-2
6
Fernando Dı́az-Espadaa,†, Victor Matheub, and Yvelise Barriosc,*
a
Department of Immunology, Clı´nica Puerta de Hierro, Madrid, Spain
b
Servicio de Alergologı´a, Floor-2, Edificio de Actividades Ambulatorias, Hospital Universitario de
Canarias, Tenerife, Spain
c
Laboratorio Immunologı´a Central Lab, Planta 0, Edificio Principal, Hospital Universitario de
Canarias, Tenerife, Spain
*Corresponding author: e-mail address: [email protected]
Abbreviations
ACE2 angiotensin-converting enzyme 2
ADCC antibody mediated cellular cytotoxicity
AGEP acute generalized exanthematous pustulosis
BCR B-cell receptor
BAT basophil activation test
CMV cytomegalovirus
COVID-19 coronavirus disease 2019
CoviDCELL® register name of Spike DTH
DTH delayed-type hypersensitivity
DRESS drug reaction with eosinophilia and systemic symptoms
E envelope structural protein of SARS-CoV-2
EIA enzyme immunoassays
FDE fixed drug exanthema
G&C Gell & Coombs
HLA human leukocyte antigen
HCoV human coronavirus
IDT intradermal tests
IGRA interferon gamma release assays
†
RETIRED
1 Historical perspective
The expression “delayed type hypersensitivity” was introduced in the immunologi-
cal vocabulary by the British immunologists Philip Gell and Robert Coombs (Gell &
Coombs, 1963), in their seminal classification that categorized hypersensitivity dis-
eases of immune origin into four classes (Table 1), according to their particular ef-
fector mechanisms. In its original meaning, the word “hypersensitivity” denoted the
status of a mammalian organism immunized against a microbial pathogen and its
ability to react against it after a new exposure to the same agent. But the enormous
size of the repertoire expressed by the adaptive immune system enables the recog-
nition of an astonishing number of antigenic structures, that extend well beyond the
substances present in infectious (microbial) and non-infectious (ectoparasites, chem-
ical and toxic compounds) agents, and include molecules present in the host tissues
(tumour cells, autoantigens). Although the immune system machinery is under a
strict control regime, subtle alterations in its functioning can result in exaggerated
reactions that may cause damage to the tissues of the host. As the 1960s progressed,
the dysfunction of the immune system was increasingly recognized as a pathogenic
mechanism, counteracting the previous appreciation, firmly rooted in the first few
decades of the 20th century, of its beneficial effects in the prevention and resolution
of infectious diseases. In the context in which G&C gave birth to their celebrated
classification, the word hypersensitivity was employed to describe exclusively the
Table 1 Classification of hypersensitivity reactions.
Type II Type IV
Latency Immediate (seconds Min to hours Min to hours Up to 12 h 12 h to days 12 h to days 12 h to days 12 h to days
to minutes)
Immune IgE IgG/IgM IgG/IgM IgG/IgM CD4 + TH1 CD4 + TH2 CD8 + CD4 + TH17
reactant cells cells T cells cells
Antigens Eukaryotic antigens Cell Cell Soluble antigens Intracellular Parasitic Virus Medications
Foreign/self enzymes, toxins, membrane membrane Bacterial antigens bacteria, virus worms Medications (haptens, p-i)
venoms, xenobiotics molecules molecules Viral particles Medications Chemicals
Drugs acting as Extracellular Antibody (haptens, (haptens,
haptens matrix pharmaceuticals p-i) p-i)
Drugs (hapt)
Effector Mast cell C0 deposition Interference Immunocomplex Macrophage Eosinophilic Cytotoxic Neutrophilic
mechanism degranulation IgE Phagocytosis with cell C0 deposition, activation inflammation infiltration
mediated ADCC function PMN influx. Granuloma
Histamine, PMN influx formation
leukotrienes
Beneficial Parasitic expulsion, Extracellular Clearance bacterial Control Granuloma Virus Enhance
Reactions toxin removal: bacteria lysis. antigens and viral Mycobacterial eggs from removal phagocytosis
increased peristalsis, particles infection helminths bacteria/virus
mucous secretion,
edema, diarrhoea
Detrimental Allergic diseases, MBT/Rh/HA Myasthenia Serum sickness Insulitis DRESS SJS/TEN Pustular
reactions anaphylaxis Autoimmune gravis, Graves Arthus reaction syndrome Contact psoriasis
hypothyroidis, disease, SLE, reactive dermatitis DAGEP
Good Pasture Chronic arthritis,
Pemphigus idiopathic polyarteritis
Rheumatic urticaria nodosa, allergic
fever alveolitis, PSGN
ADCC, antibody dependent cellular cytotoxicity; DAGEP, drug induced acute generalized exanthematous pustulosis; DRESS: drug reaction with eosinophilia and systemic symptoms; HA,
hemolytic anaemia; MBT, mismatched blood transfusion; PSGN, post-streptococcal glomerulonephritis; Rh, rhesus incompatibility; SJS/TEN, Stevens-Johnson syndrome/toxic epidermal
necrolysis; SLE, systemic lupus erythematosus.
192 CHAPTER 6 DTH methods and immune response to covid
harmful reactions that occurred during immune responses. In an effort to avoid its
ambiguous meaning, G&C describe these pathogenic responses as “allergic reactions
producing tissue damage”.
The G&C classification divides hypersensitivity reactions into four pathophysi-
ological categories. The first three types describe reactions conveyed by antibodies
and are considered “immediate” because its manifestations occur within the first 24 h
after the initial triggering event, whereas the fourth type described hypersensitivity
reactions accomplished by the T cell arm of the immune system and is considered
“delayed” because the reactions are not seen until 24–48 h. The great amount of ad-
vances in our understanding of the functioning of the immune system since the year
of G&C report, have led to a re-interpretation of their classification. While types
I and III have remained unchanged since G&C devised their classification of hyper-
sensitivity reactions, types II and IV have been subclassified in two and four sub-
types, respectively, and a fifth type of hypersensitivity reaction has been proposed
to accommodate sarcoid diseases. Despite these advances, we think that the simple
G&C classification has withstood the test of time reasonably well and is still widely
used to describe the pathologies resulting from unwanted reactions of the immune
responses.
But it must be considered that the system devised by G&C to categorize delete-
rious reactions induced by rather innocuous substances can be easily applied to cat-
egorize beneficial reactions used by the immune system that allows the host to get rid
of microbial invaders. In this regard, in vivo tests based on the G&C principles are
currently used not only to diagnose patients who have experienced hypersensitivity
reactions to certain substances but also to investigate the immune status of individ-
uals affected by a particular microbial infection. Both the skin tests to demonstrate
immune reaction to certain substances (contact dermatitis) and the tuberculin reac-
tion are both diagnostic procedures contemplated as G&C Type IV reactions that re-
veal either a pathological hypersensitivity event or a normal immune response to a
past microbial infection. Well before the discovery of T cell recirculation and the
existence of skin resident memory cells, Richard Wagner wrote, in his emotive hom-
age to the figure of Clemens von Pirquet, “Out of the darkness of inner parts of the
body and submerged tissues, the pathological processes and reactions were
projected onto the surface and moved into bright light” (Wagner, 1964). Today’s
immunologists take advantage of this projection and have in their hands a simple
and affordable method to investigate the immune reaction to a wide variety of
microbial and non-microbial challenges.
It is important to take into account that some clinical symptoms may overlap
among the different classes of hypersensitivity and that many small molecular weight
drugs can cause all types of hypersensitivity reactions, mostly involving type I or
type IV hypersensitivity reactions. Adverse reactions to drugs (medications) are par-
ticularly important in clinical practice (Edwards & Aronson, 2000) and were defined
by the World Health Organization (WHO) in 1972 as “a response to a drug which is
noxious and unintended, and which occurs at doses normally used in humans for the
prophylaxis, diagnosis, or therapy of disease, or for the modification of physiological
function”.
2 Hypersensitivity reactions 193
skin tests. Skin tests and measurements of specific IgE antibodies in serum give com-
plementary information for the diagnosis of allergic diseases.
When the skin test or the specific IgE immunoassays are not conclusive,
the basophil activation test (BAT), that measures either histamine release
(Ostergaard, Ebbensen, Nolte, & Skov, 1990) or CD63 upregulation (González-
Muñoz, Villota, & Moneo, 2008) following stimulation of blood basophiles with
allergen in vitro, emerged as a new diagnostic tool. The test tries to reproduce
in vitro the allergic reaction in patients sensitized to particular allergens. Both
EIA and BAT studies should be used in patients in whom skin challenge can cause
reactions of unpredictable severity.
complex (C5b-C9). However, this lytic mechanism may not be very efficient, due to
the phenomenon of homologous restriction, whereby cells are protected from lysis by
autologous complement by a self-recognition mechanism that inhibits the late phases
of complement attack, composed of the cell membrane proteins protectin (CD59),
cofactor protein MCP (CD46) and decay-accelerating factor DAF (CD55) (Gorter
et al., 1996). Nevertheless, the first steps of complement fixation results in the de-
position and covalent binding of iC3b to the cell membrane of blood cells, which
renders these cells susceptible to phagocytosis by specialized macrophages in the
liver and spleen that express the iC3b complement receptors CR3 (CD11b/CD18)
and CR4 (CD11c/CD18) (Takizawa, Tsuji, & Nagasawa, 1996). This particular
way of cell destruction links complement fixation to phagocytosis, and enhances
the deleterious effects (anaemia, neutropenia or thrombocytopenia) of type IIa hy-
persensitivity reactions. Moreover, complement fixation on target cells results in
the local production of anaphylotoxins (C3a, C5a) that recruit polymorphonuclear
leukocytes (Forema, Glovsky, Warner, Horvath, & Ward, 1996) and amplify tissue
injury through the release of hydrolytic enzymes after their autolysis.
Type IIa hypersensitivity is typified by the reactions of preformed antibodies in
mismatched blood transfusion (Davenport & Mintz, 2007) and in the haemolytic dis-
ease of the newborn (Rhesus incompatibility) (Murray & Roberts, 2007), which lead
to alloimmune destruction of red blood cells. The different types of autoimmune de-
struction of blood cells (autoimmune haemolytic anaemia, ANCA-dependent neutro-
penias, idiopathic thrombocytopenic purpura) and organ-specific autoimmune
diseases (autoimmune hypothyroidisms, anti-GBM nephritis and Goodpasture syn-
drome, pemphigus vulgaris) are also examples of type IIa reactions in which anti-
bodies directed against self-antigens promote the destruction of cells and tissues
(Kumar, Abbas, & Aster, 2021). A special case of type IIa hypersensitivity is rheu-
matic fever, in which epitope similarity between streptococcal antigens and myocar-
dial or brain antigens may explain the presence of reacting antibodies that contribute
to pathologies affecting these organs (Cunningham et al., 1989; Guilherme, Kalil, &
Cunningham, 2006).
Although drug hypersensitivity reactions are most frequently mediated by IgE
(Type I) or T cells (type IV), they can also induce anaemia, neutropenia, thrombocy-
topenia and hepatic dysfunction, all hallmarks of IgG mediated, type IIa hypersensi-
tivity reactions. To become immunogenic, drugs act as haptens, requiring conjugation
to a cell surface protein on blood cells or other carrier proteins. Antibiotics (penicillins,
cephalosporins), antihypertensive drugs (alpha methyldopa), thiazides, quinidine and
other drugs can induce haemolytic anaemia and thrombocytopenic purpura through
this mechanism (Kaufman et al., 1993; Petz, 1993).
In type IIb hypersensitivity, autoantibodies bind to receptors on the target cells,
inducing dysfunction of the affected organ. In Graves’ disease, anti-thyrotropin re-
ceptor antibodies act as agonists and stimulate the thyroid gland to produce excessive
amounts of thyroid hormone (hyperthyroidism) (Chen et al., 2003). In some cases of
chronic idiopathic urticaria, FcεRI on mast cells are recognized by auto IgG anti-
bodies, which causes the degranulation of the target cells and the onset of urticaria
198 CHAPTER 6 DTH methods and immune response to covid
complex is a double-edged sword. On the one hand, the iC3b deposited in immuno-
complexes facilitates solubilisation (Whaley & Ahmed, 1989) or removal through
interaction with CR1 receptors on phagocytic cells in the liver (Katyal,
Sivasankar, & Das, 2001). Although it might seem counter-intuitive, another bene-
ficial effect of C3b deposition is the disaggregation of immune complexes in smaller
entities (Miller & Nussenzweig, 1975) that can be more easily engulfed by phago-
cytic cells (Petersen, Baatrup, Jepsen, & Svehag, 1985). An important component of
humoral anti-viral responses might be the reactions initiated by complement depo-
sition on antigen–antibody complexes, that can contribute to the elimination of an-
tibody coated circulating viral particles (Rajan, 2003). In this context, the C3b is said
to have “neutralized” the virus.
But on the other hand, anaphylatoxins (C3a, C5a) liberated during the early phase
of complement fixation attract and activate polymorphonuclear leucocytes
(Mayadas, Tsokos, & Tsuboi, 2009) which release mediators that cause inflamma-
tory damage to the tissues. Anaphylatoxins can also activate local mast cells, induc-
ing their degranulation and the release of mediators that increase vasodilatation and
vasopermeability. Depending on the site of deposition, symptoms of vasculitis (of
endothelial cells of blood vessels), purpuric rash (dermis), arthritis (joints) or glo-
merulonephritis (renal glomeruli) can develop. In the case of inhalational entry,
hypersensitivity pneumonitis (allergic alveolitis) can ensue. The “farmer’s lung”
disease (Campbell, 1932) is a potentially dangerous hypersensitivity pneumonitis
that has attained considerable importance in respiratory medicine, and afflicts agri-
cultural workers exposed to organic material such as dust of improperly dried grains
or spores of fungus that grow in certain crops.
Hypersensitivity III reactions are implicated in a number of autoimmune, infec-
tious or drug-induced diseases. Systemic lupus erythematosus (SLE), a chronic,
autoimmune inflammatory disorder of connective tissue, is a prototypic type III
hypersensitivity disease, in which antibodies against nuclear components react with
released chromatin from apoptotic debris forming immune complexes. In the more
severe forms of SLE, these immune complexes can deposit in different organs, lead-
ing to a wide variety of abnormalities including nephritis, arthritis, cutaneous rash
(small vessels) and mesenteric vasculitis or mononeuritis (medium to large size
vessels) (Aranow, Diamond, & Mackay, 2008).
Infectious diseases like hepatitis B, bacterial endocarditis and yersiniosis display
a continuous source of antigens to form circulating immune complexes, causing
polyarteritis nodosa, poststreptococcal glomerulonephritis and reactive arthritis,
respectively (Kumar et al., 2021).
At the current time, serum sickness is associated with medications containing het-
erologous proteins (snake antivenom immunoglobulins, anti-thymocyte globulin,
protein vaccines, thrombolytic therapies, chimeric monoclonal antibodies) or with
insect stings. The local injection of the antigen may cause a necrotizing skin lesion
(Arthus reaction). Repeated transfusions containing residual amounts of plasma or
infusion of normal plasma in IgA deficient patients may induce anaphylactoid reac-
tions suggestive of type III hypersensitivity. Despite the wide use of medications
200 CHAPTER 6 DTH methods and immune response to covid
containing potential inducers of type III reactions, the annual rate of serum sickness
incidence is low (Rixe & Tavarez, 2021).
damage to the host tissues. Moreover, the adverse reactions to some drugs and che-
micals also have notorious delayed type hypersensitivity manifestations, and are
even more common than immediate type I allergic reactions and in some cases they
can be life-threatening. Many drugs or chemicals are capable of eliciting type IV re-
actions, including beta-lactam antibiotics, quinolones, tetracyclines, certain NSAIDs
like oxicam, X-ray contrast media, metallic ions, poison ivy, local anaesthetics, al-
lopurinol, anticonvulsants, anticoagulants like LMW heparins, and antiviral and
antifungal medications (Brandt & Bircher, 2017).
The covalent binding of drugs to self-proteins (hapten–carrier complex) can eas-
ily explain antibody-based hypersensitivity reactions, and the same mechanism can
be involved in some cases of delayed type hypersensitivity reactions to medical
drugs. Beta-lactam antibiotics can form covalent links with peptides bound in the
groove of surface HLA molecules or react with self-proteins, that after intracellular
processing, yield hapten modified peptides, giving rise to new T cell epitopes
(Weltzien & Padovan, 1998). But in some cases, drugs and other chemicals that lack
hapten characteristics bind noncovalently to the immune receptors involved in T cell
activation, promoting the polyclonal or oligoclonal stimulation of T cells. These
compounds that modified T cell reactivity are of great medical relevance. Unlike
conventional type IV hypersensitivity, the induction of reactions by the pharmaco-
logical interaction of drugs with immune receptors (p-i concept, Pichler, 2008) does
not require a previous drug-specific sensitization. In some cases (sulphamethoxa-
sole), the drug alters the T cell receptor directly, initiating a signalling event that
must be completed by conventional TCR-HLA interactions. In others (abacavir,
carbamazepine), the drugs interact with peptide-binding pockets of certain HLA
alleles inducing changes in the shape of bound self-peptides (Ramsbottom, Carr,
Jones, & Rigden, 2018). This seems to be sufficient to create neoepitopes that trigger
unwanted T cell responses, probably involving preactivated T cells. Abacavir, an an-
tiretroviral agent indicated for the treatment of HIV-1 infection, binds to the peptide
cleft of the allele B*5701, and induces a severe and systemic hypersensitivity
reaction, precluding its use in patients expressing that allele (Mallal et al., 2002).
See Fig. 1 for a more detailed description of drug-induced type IV hypersensitivity
reactions.
Naı̈ve T lymphocytes differentiate into distinct subpopulations depending on the
nature of the invading antigen, the type of presenting cell and the cytokine microen-
vironment. Depending on the T cell subpopulation involved, type IV reactions can be
further subdivided into Ia, IVb, IVc and IVd subtypes. Each of the distinct
T phenotypes release certain chemokines and cytokines that preferentially recruit
and activate monocytes (type IVa), eosinophils (type IVb), or neutrophils (type
IVd). In type IVc reactions cytotoxic T lymphocytes participate in the direct killing
of target cells.
Type IVa reactions are mediated by the TH1 lymphocyte subset. A prototypical
example is the tuberculin reaction, characterized by a preferential TH1-type cytokine
profile with significant increases in the numbers of IL-2 and IFN-gamma mRNA-
expressing cells (Tsicopoulos et al., 1992) and activation of macrophages.
202 CHAPTER 6 DTH methods and immune response to covid
FIG. 1
Models of drug interactions with the T cell receptor/peptide/HLA complex that modify T cell
reactivity. A normal event in T cell recognition is depicted in A). A processed peptide (only five
aa are shown for simplicity) fits into the floor of the binding cleft of a HLA molecule. Typically,
two amino acid side chains bind into particular anchor pockets (p) in the base of the groove.
A TCR paratope is locked into the topside of the peptide. The trimolecular complex is further
stabilized by interactions between the TCRαβ chains and the α helix domains of the HLA
molecule (). In the hapten model (B), a drug/chemical (e.g., penicillin, ) binds covalently to a
self-protein that is processed by an APC and presented as short peptides, some of which can
bear the haptenized fragment (neoepitope). If recognized by the TCR of a non-tolerized T cell,
an unpredicted immune response can ensue. In (C), (p-i TCR concept) a drug/chemical like
sulphamethoxazole () bind non-covalently to the paratope of a TCR, and alters its
conformation inducing a stimulatory signal regardless of the bound peptide, that is
complemented by the canonical TCR-HLA interactions. The reaction is similar to that seen in
an alloimmune response. Alternatively (D), the drug (e.g., abacavir) () can bind non-
covalently to the binding pocket of an HLA molecule (p-i HLA concept), altering the shape of
the permissible peptides without the requirement of intracellular processing or allowing the
attachment of a different array of peptides. The new epitopes can induce a polyclonal
activation of T cells. The p-i-HLA concept has also been called “altered peptide repertoire
model”.
2 Hypersensitivity reactions 203
When the patients have suffered non-immediate reactions and the suspect path-
ogenic mechanism is a T-cell mediated hypersensitivity (Lerch & Pichler, 2004;
Rozieres, Vocanson, Saı̈d, Nosbaum, & Nicolas, 2009), skin IDT is also useful
(Joshi & Khan, 2021), but more controversial (Phillips et al., 2019). In those cases,
the late reading of IDT can be delayed from 12 h to several days later (Romano et al.,
2004). In some diseases such as acute generalized exanthematous pustulosis
(AGEP), Stevens-Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN)
(Iriki et al., 2014), the reading of the skin tests must be done at 24–48 h or even
1 week after IDT. In these disorders, the presence of effector-memory T cells and
intraepidermal CD8(+) T cells with the local production of interferon gamma after
the introduction of the triggering agent (Mizukawa et al., 2002) but also the presence
of skin resident memory T cells have been shown (Tokura et al., 2021).
Patch tests are diagnostic tools mainly in contact dermatitis. But also, non-
immediate drug reactions like those previously named AGEP, SJS and TEN, drug
reactions with eosinophilia and systemic symptoms (DRESS) (Cabañas et al., 2020),
single organ diseases and fixed drug exanthema (FDE) (Patel, John, Handler, &
Schwartz, 2020), could also benefit from the use of a skin patch test for effective diag-
nosis (Copaescu, Gibson, Li, Trubiano, & Phillips, 2021). In all those conditions, a late
reading of at least 48 h or more (Bhujoo et al., 2021) would give us a picture similar to
that seen in contact dermatitis and would approximate a true etiological diagnosis
(Belsito, 1989) with T cell involvement (Adam, Pichler, & Yerly, 2011).
different available methods, because differences in the published results could cor-
respond to methodological details. In conclusion, IGRAs are an important diagnostic
tool in COVID infected and vaccinated individuals but, in order to expand its use,
some uncertainties about the cut-off values and the real-life correspondence with
protection from infection needs to be further defined to augment the prognostic value
of a positive IGRA result.
As explained before, the Mantoux tuberculin skin test (TST) was developed early
in the 20th century, and although with some drawbacks, it is still present in the actual
medical practice (Richeldi, 2009). During the last few decades it has been used in
parallel with the IGRA tests, producing an enormous amount of data permitting
the comparison of both methods (Hass & Belknap, 2019). All these data have been
used to improve the diagnosis of latent tuberculosis infection, where the performance
of IGRA tests have been demonstrated to be superior in terms of discriminating in-
fection in vaccinated individuals (Lewinsohn et al., 2017). This has been achieved
using IGRA tests with peptides not present in the vaccine formulation that are not
possible to distinguish in the TST test.
However, the analysis of the situation in SARS-CoV-2 infection is completely
different. In these COVID patients the focus of the results should be the sensitivity
of the test, finding as many individuals as possible that either by natural exposure or
after vaccination could have developed an adaptive T cell response. In that scenario
an in vivo presentation of the antigen to the T-cell population could be superior if
these two methods are compared (see Table 2 to understand differences exhibited
between IGRA and TST).
FIG. 2
Upper: example of response to Candida (C) antigen after intradermal test in a subject
at 12, 24, 48 and 72 h. Below: example of response to Spike (S) antigen after intradermal test
in a subject with a fully recovered SARS-CoV-2 disease at 12, 24, 48 and 72 h.
patients into different groups depending on their clinical presentation. From this set
of tests, it was evident that DTH using RBD derived S protein from SARS-CoV-2 is a
simple method to investigate immunity after virus exposure. The DTH test that was
performed in a group of non-exposed individuals was negative in all of the cases,
showing a high specificity of the test. Moreover, comparisons between specific
anti-RBD IgG and spike-DTH cutaneous test results, to identify the exposed individ-
uals, showed a concordance number of 84.3%. In this set of exposed individuals,
CoviDCELL® showed a superior capacity to identify exposed individuals.
Although until this manuscript has been written, there were no more scientific
public data regarding the use of DTH to assess immune exposure to SARS-CoV-2,
several biotech companies have announced the use of peptide-base or protein-
derived moieties from different parts of the virus with the intended use of developing
commercial delayed type hypersensitivity tests to assess T-cell immunity after infec-
tion or to measure immunogenicity elicited by the vaccines. One of the concerns re-
garding the anti-COVID-19 DTH reaction is the performance of the test with the new
variants produced by the virus. In this regard, ELISAs can be more susceptible to
changes in the conformational epitopes recognized by the patient’s antibodies.
But T-cells recognize many linear epitopes, and a substantial number of changes
in the potential epitopes between the variants would be required to affect the
T-cell recognition. These facts suggest that the skin test will be relatively unaffected
by the virus variants.
6 DTH to measure immunogenicity elicited by covid vaccines 213
FIG. 3
Upper: example of response to Spike (S) antigen after intradermal test at 12, 24, 48 and 72 h
in a fully vaccinated subject with a mRNA vaccine. Below: example of response to Spike
(S) antigen after intradermal test at 12, 24, 48 and 72 h in a subject with a fully recovered
SARS-CoV-2 disease and two doses of a mRNA vaccine.
214 CHAPTER 6 DTH methods and immune response to covid
Acknowledgements
To the patients and participants in the DTH-studies.
To our collaborators (Dr. Franco and Dra. Alava) and technical assistants for their contin-
ued help and support.
Victor Matheu and Yvelise Barrios have registered (50%) CoviDCELL® 202199800
402351.
References
Adam, J., Pichler, W. J., & Yerly, D. (2011). Delayed drug hypersensitivity: Models of T-cell
stimulation. British Journal of Clinical Pharmacology, 71, 701–707.
Adler, N. R., Aung, A. K., Ergen, E. N., Trubiano, J., Goh, M. S. Y., & Phillips, E. J. (2017).
Recent advances in the understanding of severe cutaneous adverse reactions. British
Journal Dermatology, 177, 1234–1247.
Aiello, A., Fard, S. N., Petruccioli, E., Petrone, L., Vanini, V., Farroni, C., et al. (2021). Spike
is the most recognized antigen in the whole-blood platform in both acute and convalescent
COVID-19 patients. International Journal of Infectious Diseases, 106, 338–347.
References 215
Ansotegui, I. J., Melioli, G., Canonica, G. W., Caraballo, L., Villa, E., Ebisawa, M., et al.
(2020). IgE allergy diagnostics and other relevant tests in allergy, a World Allergy
Organization position paper. World Allergy Organization Journal, 13, 100080.
Aranow, C., Diamond, B., & Mackay, M. (2008). Systemic lupus erythematosus in clinical
immunology. Principles and practice. In R. R. Rich, T. A. Fleisher, W. T.
Shearer, H. W. Schroeder Jr.,, A. J. Frew, & C. M. Weyand (Eds.), Clinical immunology.
Principles and practice. Philadelphia: Elsevier (Chapter 51).
Arthus, M. (1903). Injections repetees de serum du cheval chez le lapin. Comptes rendus des
s
eances de la Soci et
e de biologie et de ses filiales, 55, 817–820.
Badaro, R., Machado, B. A. S., Duthie, M. S., Araujo-Neto, C. A., Pedral-Sampaio, D.,
Nakatani, M., et al. (2020). The single recombinant M. tuberculosis protein DPPD
provides enhanced performance of skin testing among HIV-infected tuberculosis patients.
AMB Express, 10, 133.
Barrios, Y., Franco, A., Alonso-Larruga, A., Sánchez-Machı́n, I., Poza-Guedes, P.,
Gonzalez, R., et al. (2020). Success with multidisciplinary team work: Experience of a
primary immunodeficiency unit. Journal of Investigational Allergology and Clinical Im-
munology, 30, 208–210.
Barrios, Y., Franco, A., Sanchez-Machin, I., Poza-Guedes, P., Gonzalez-Perez, R., &
Matheu, V. (2021). A novel application of delayed-type hypersensitivity reaction to
measure cellular immune response in SARS-CoV-2 exposed individuals. Clinical Immu-
nology, 226, 108730.
Barrios, Y., Franco, A., Sánchez-Machı́n, I., Poza-Guedes, P., González-Perez, R., &
Matheu, V. (2021). The beauty of simplicity: Delayed-type hypersensitivity reaction to
measure cellular immune responses in RNA-SARS-Cov-2 vaccinated individuals. Vac-
cines (Basel), 9, 575. https://doi.org/10.3390/vaccines906057.
Baryaliya, M., Sanmukhani, J., Patel, T., Paliwal, N., Shah, H., & Tripathi, C. (2011).
Drug-induced Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), and
SJS-TEN overlap: A multicentric retrospective study. Journal of Postgraduate Medicine,
57, 115–119.
Baumgarth, N., Nikolich-Žugich, J., Lee, F. E., & Bhattacharya, D. (2020). Antibody
responses to SARS-CoV-2: Let’s stick to known knowns. Journal of Immunology, 205,
2342–2350.
Belsito, D. V. (1989). The immunologic basis of patch testing. Journal of the American Acad-
emy of Dermatology, 21, 822–829.
Bennich, H. H., Ishizaka, K., Johansson, S. G. O., Rowe, D. S., Stanworth, D. R., &
Terry, W. D. (1968). Immunoglobulin IgE, a new class of human immunoglobulin.
Bulletin World Health Organization, 38, 151–152.
Berot, V., Gener, G., Ingen-Housz-Oro, S., Gaudin, O., Paul, M., Chosidow, O., et al. (2020).
Cross-reactivity in beta-lactams after a non-immediate cutaneous adverse reaction:
Experience of a reference centre for toxic bullous diseases and severe cutaneous adverse
reactions. Journal of the European Academy of Dermatology and Venereology, 34, 787–794.
Bhujoo, Z., Ingen-Housz-Oro, S., Gener, G., Gaudin, O., Fleck, M., Verlinde-Carvalho, M.,
et al. (2021). Patch tests in nonimmediate cutaneous adverse drug reactions: The impor-
tance of late readings on day 4. Contact Dermatitis. https://doi.org/10.1111/cod.13981.
Bocquet, H., Bagot, M., & Roujeau, J. C. (1996). Drug-induced pseudolymphoma and drug
hypersensitivity syndrome (Drug Rash with Eosinophilia and Systemic Symptoms:
DRESS). Seminars in Cutaneous Medicine and Surgery, 15, 250–257.
Brandt, O., & Bircher, A. J. (2017). Delayed-type hypersensitivity to oral and parenteral drugs.
Journal der Deutschen Dermatologischen, 15, 1111–1132.
216 CHAPTER 6 DTH methods and immune response to covid
Burrell, C. J., Howard, C. R., & Murphy, F. A. (2016). Fenner and White’s medical virology
(5th ed.). United States: Academic Press.
Cabañas, R., Ramı́rez, E., Sendagorta, E., Alamar, R., Barranco, R., Blanca-López, N., et al.
(2020). Spanish guidelines for diagnosis, management, treatment, and prevention of
DRESS syndrome. Journal of Investigational Allergology and Clinical Immunology,
30, 229–253.
Caiado, J., Venemalm, L., Pereira-Santos, M. C., Costa, L., Barbosa, M. P., & Castells, M.
(2013). Carboplatin-, oxaliplatin-, and cisplatin-specific IgE: Cross-reactivity and value
in the diagnosis of carboplatin and oxaliplatin allergy. The Journal of Allergy and Clinical
Immunology. In practice, 1, 494–500.
Campbell, J. M. (1932). Acute symptoms following work with hay. British Medical Journal, 2,
143–144.
Castells, M. (2018). New role for the modern allergist in drug allergy: Assess, diagnose, and
de-label. Annals of Allergy, Asthma & Immunology, 121, 515–516.
Chase, M. (1961). The preparation and standardization of Kveim testing antigen. American
Review of Respiratory Disease, 84, 86–88.
Chen, C.-R., Pichurin, P., Nagayama, Y., Latropha, F., Rapaport, B., & McLachlan, S. M.
(2003). The thyrotropin receptor autoantigen in Graves disease is the culprit as well as
the victim. Journal of Clinical Investigation, 111, 1897–1904.
Clark, R. A., Chong, B., Mirchandani, N., Brinster, N. K., Yamanaka, K., Dowgiert, R. K.,
et al. (2006). The vast majority of CLA + T cells are resident in normal skin. Journal
of Immunology, 176, 4431–4439.
Coca, A. F., & Grove, E. F. (1923). A study of the atopic reagin. Proceedings of the Society for
Experimental Biology and Medicine, 21, 49.
Copaescu, A., Gibson, A., Li, Y., Trubiano, J. A., & Phillips, E. J. (2021). An updated review
of the diagnostic methods in delayed drug hypersensitivity. Frontiers in Pharmacology.
https://doi.org/10.3389/fphar.2020.573573.
Cunningham, M. W., McCormack, J. M., Fenderson, P. G., Ho, M. K., Beachey, E. H., &
Dale, J. B. (1989). Human and murine antibodies cross-reactive with streptococcal
M protein and myosin recognize the sequence GLN-LYS-SER-LYS-GLN in M protein.
Journal of Immunology, 143, 2677–2683.
Dammermann, W., Bentzien, F., Stiel, E. M., K€ uhne, C., Ullrich, S., Schulze Zur Wiesch, J.,
et al. (2015). Development of a novel IGRA assay to test T cell responsiveness to HBV
antigens in whole blood of chronic Hepatitis B patients. Journal of Translational
Medicine, 13, 157.
Dan, J. M., Mateus, J., Kato, Y., Hastie, K. M., Yu, E. D., Faliti, C. E., et al. (2021). Immu-
nological memory to SARSCoV-2 assessed for up to 8 months after infection. Science
(New York, N.K.), 371(6529), eabf4063. https://doi.org/10.1126/science.abf4063.
Davenport, R. D., & Mintz, P. D. (2007). Transfusion medicine. In R. A. McPherson, & M. R.
Pincus (Eds.), Henry’s clinical diagnosis and management by laboratory methods
(pp. 669–684). Philadelphia: Saunders Elsevier.
De Vries, J. E., Punnonen, J., Cocks, B. G., de Waal Malefyt, R., & Aversa, G. (1993). Regu-
lation of the human IgE response by IL4 and IL13. Research in Immunology, 144, 597–601.
Del Prete, G. (1992). Human Th1 and Th2 lymphocytes: Their role in the pathophysiology of
atopy. Allergy, 47, 450–455.
Demoly, P., Romano, A., & Bousquet, J. (2008). In vivo methods for the study of allergy.
In N. F. Adkinson, J. W. Yunginger, W. W. Busse, B. S. Bochner, F. E. R. Simons, &
S. T. Holgate (Eds.), Middleton’s allergy: Principles and practice (pp. 1267–1280).
Amsterdam: Mosby, Inc.
References 217
Joshi, S. R., & Khan, D. A. (2021). Non-IgE-mediated drug hypersensitivity reactions. Current
Allergy and Asthma Reports, 31, 41.
Katyal, M., Sivasankar, B., & Das, N. (2001). Complement receptor 1 in autoimmune disor-
ders. Current Science, 81, 907–914.
Kaufman, D. W., Kelley, J. P., Johannes, C. B., Sandler, A., Harmon, D., Stolley, P. D., et al.
(1993). Acute thrombocytopenic purpura in relation to the use of drugs. Blood, 82,
2714–2718.
Kim, S.-H. (2020). Interferon-γ release assay for cytomegalovirus (IGRA-CMV) for risk
stratification of posttransplant CMV infection: Is it time to apply IGRA-CMV in routine
clinical practice? Clinical Infectious Diseases, 71, 2386–2388.
Koch, R. (1890). A remedy for tuberculosis. Lancet, 136, 1085–1086.
Kramer, A., Schwebke, I., & Kampf, G. (2006). How long do nosocomial pathogens persist on
inanimate surfaces? A systematic review. BMC Infectious Diseases, 6, 130.
Kumar, V., Abbas, A. K., & Aster, J. C. (2021). Robbins & Cotran, pathologic basis of disease
(Chapter 6). Philadelphia: Elsevier.
Landsteiner, K. (1945). The specificity of serological reactions. Cambridge, MA: Harvard
University Press.
Landsteiner, K., & Chase, M. W. (1942). Experiments on transfer of cutaneous sensitivity to
simple compounds. Proceedings of the Society for Experimental Biology, 49, 688–690.
Lerch, M., & Pichler, W. J. (2004). The immunological and clinical spectrum of delayed drug-
induced exanthems. Current Opinion in Allergy and Clinical Immunology, 4, 411–419.
Lewinsohn, D. M., Leonard, M. K., LoBUe, P. A., Cohn, D. L., Daley, C. L., Desmond, E.,
et al. (2017). Official American Thoracic Society/infectious diseases. Society of America/
Centers for disease control and prevention. Clinical practice guidelines: Diagnosis of
tuberculosis in adults and children. Clinical Infectious Diseases, 64, 111–115.
Li, J., & Uetrecht, J. P. (2010). The danger hypothesis applied to idiosyncratic drug reactions.
Handbook of Experimental Pharmacology, 196, 493–509.
Liu, Y. J. (2006). Thymic stromal lymphopoietin: Master switch for allergic inflammation.
Journal of Experimental Medicine, 203, 269–273.
Liu, D. X., Fung, T. S., Chong, K. K., Shukla, A., & Hilgenfeld, R. (2014). Accessory proteins
of SARS-CoV and other coronaviruses. Antiviral Research, 109, 97–109.
Liu, W. J., Zhao, M., Liu, K., Xu, K., Wong, G., Tan, W., et al. (2017). T-cell immunity of
SARS-CoV: Implications for vaccine development against MERS-CoV. Antiviral Re-
search, 137, 82–92.
Lochmatter, P., Zawodniak, A., & Pichler, J. (2009). In vitro tests in drug hypersensitivity
diagnosis. Immunology Allergy Clinics of North America, 29, 537–554.
Lu, R., Zhao, X., Li, J., Niu, P., Yang, B., & Wu, H. (2020). Genomic characterisation and
epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor
binding. Lancet, 395, 565–574.
Mahmoudi, S., Mamishi, S., Suo, X., & Keshavarz, H. (2017). Early detection of Toxoplasma
gondii infection by using an interferon gamma release assay: A review. Experimental
Parasitology, 172, 39–43.
Mallal, S., Nolan, D., Witt, C., Masel, G., Martin, A. M., Moore, C., et al. (2002). Association
between presence of HLA-B*5701, HLA-DR7, and HLA-DQ3 and hypersensitivity to
HIV-1 reverse-transcriptase inhibitor abacavir. Lancet, 359, 727–732.
Mantoux, C. (1908). Intradermo-reaction de la tuberculine. Comptes rendus de l’Acad emie des
sciences, Paris, 147, 355–357.
References 219
Martı́nez-Gallo, M., Esperalba, J., Pujol-Borrell, R., Sandá, V., Arrese-Muñoz, I., Fernández-
Naval, C., et al. (2021). Commercialized kits to assess T-cell responses against
SARS-CoV-2 S peptides. A pilot study in health care workers. Medicina Clinica
(Barc). https://doi.org/10.1016/j.medcli.2021.09.013. S0025-7753(21)00529-7. English,
Spanish.
Mayadas, T. N., Tsokos, G. C., & Tsuboi, N. (2009). Mechanisms of immune complex-
mediated neutrophil recruitment and tissue injury. Circulation, 120, 2012–2024.
McKenzie, S. E., & Schreiber, A. D. (1998). Fc gamma receptors in phagocytes. Current
Opinion in Hematology, 5, 16–21.
Metzger, H., Kinet, J. P., Blank, U., Miller, L., & Ra, C. (1989). The receptor with high affinity
for IgE. Ciba Foundation Symposium, 147, 93–101.
Miller, G. W., & Nussenzweig, V. (1975). A new complement function: Solubilization of
antigen-antibody aggregates. Proceedings of the National Academy of Sciences of the
United States of America, 72, 418–422.
Mizukawa, Y., Yamazaki, Y., Teraki, Y., Hayakawa, J., Hayakawa, K., Nuriya, H., et al.
(2002). Direct evidence for interferon-gamma production by effector-memory-type intrae-
pidermal T cells residing at an effector site of immunopathology in fixed drug eruption.
The American Journal of Pathology, 161, 1337–1347.
Murphy, P. B., Atwater, A. R., & Mueller, M. (2021). Allergic contact dermatitis. In Stat-
Pearls. Treasure Island (FL): StatPearls Publishing. Available from: https://www.ncbi.
nlm.nih.gov/books/NBK532866/.
Murray, N. A., & Roberts, I. A. G. (2007). Haemolytic disease of the newborn. Archives of
Disease in Childhood, 92, 83–88.
Murugesan, K., Jagannathan, P., Pham, T. D., Pandey, S., Bonilla, H. F., Jacobson, K., et al.
(2020). Interferon-gamma release assay for accurate detection of SARS-CoV-2 T cell
response. Clinical Infectious Diseases, ciaa1537. https://doi.org/10.1093/cid/ciaa1537.
Naik, H. B., & Cowen, E. W. (2013). Autoinflammatory pustular neutrophilic diseases.
Dermatological Clinics, 31, 405–425.
Naisbitt, D. J., Hough, S. J., Gill, H. J., Pirmohamed, M., Kitteringham, N. R., & Park, B. K.
(1999). Cellular disposition of sulphamethoxazole and its metabolites: Implications for
hypersensitivity. British Journal of Pharmacology, 126, 1393–1407.
Niimi, N., Francis, D. M., Kermani, F., O’Donell, B. F., Hide, M., Kobza-Black, A., et al.
(1996). Dermal mast cell activation by autoantibodies against the high affinity IgE recep-
tor in chronic urticaria. Journal of Investigative Dermatology, 106, 1001–1006.
Ojeda, D. S., Gonzalez Lopez Ledesma, M. M., Pallares, H. M., Costa Navarro, G. S.,
Sanchez, L., Perazzi, B., et al. (2021). Emergency response for evaluating SARS-CoV-
2 immune status, seroprevalence and convalescent plasma in Argentina. PLoS Pathogens,
17, e1009161. https://doi.org/10.1371/journal.ppat.1009161.
Ostergaard, P. A., Ebbensen, F., Nolte, H., & Skov, P. S. (1990). Basophil histamine release in
the diagnosis of house dust mite and dander allergy of asthmatic children. Comparison
between prick test, RAST, basophil histamine release and bronchial provocation. Allergy,
45, 231–235.
Oyoshi, M. K., Larson, R. P., Ziegler, S. F., & Geha, R. S. (2010). Mechanical injury polarizes
skin dendritic cells to elicit a TH2 response by inducing cutaneous thymic stromal
lymphopoietin expression. Journal of Allergy and Clinical Immunology, 126, 976–984.
Palm, N. W., Rosenstein, R. K., & Medzhitov, R. (2012). Allergic host defences. Nature, 484,
465–472.
220 CHAPTER 6 DTH methods and immune response to covid
Papageorgiou, A. C., & Mohsin, I. (2020). The SARS-CoV-2 spike glycoprotein as a drug and
vaccine target: Structural insights into its complexes with ACE2 and antibodies. Cell, 9,
2343. https://doi.org/10.3390/cells9112343.
Parker, C. W. (1981). Hapten immunology and allergic reactions in humans. Arthritis and
Rheumatism, 24, 1024–1036.
Patel, S., John, A. M., Handler, M. Z., & Schwartz, R. A. (2020). Fixed drug eruptions: An
update, emphasizing the potentially lethal generalized bullous fixed drug eruption.
American Journal of Clinical Dermatology, 21, 393–399.
Pepys, J. (1975). Skin testing. British Journal of Hospital Medicine, 14, 412–416.
Peters, P. J., Borst, J., Oorschot, V., Fukuda, M., Kr€ahenb€ uhl, O., Tschopp, J., et al. (1991).
Cytotoxic T lymphocyte granules are secretory lysosomes, containing both perforin and
granzymes. The Journal of Experimental Medicine, 173, 1099–1109.
Petersen, I., Baatrup, G., Jepsen, H. H., & Svehag, S. E. (1985). Complement-mediated
solubilization of immune complexes and their interaction with complement C3 receptors.
Complement, 2, 97–110.
Petrone, L., Albrich, W. C., Tamarozzi, F., Frischknecht, M., Gomez-Morales, M. A.,
Teggi, A., et al. (2020). Species specificity preliminary evaluation of an IL-4-based test
for the differential diagnosis of human echinococcosis. Parasite Immunology, 42,
e12695. https://doi.org/10.1111/pim.12695.
Petrone, L., Petruccioli, E., Vanini, V., Cuzzi, G., Najafi Fard, S., Alonzi, T., et al. (2021).
A whole blood test to measure SARS-CoV-2-specific response in COVID-19 patients.
Clinical Microbiology and Infection, 27(2), 286.e7–286.e13. https://doi.org/10.1016/j.
cmi.2020.09.051.
Petrone, L., Vanini, V., Amicosante, M., Corpolongo, A., Gomez Morales, M. A.,
Ludovisi, A., et al. (2017). A T-cell diagnostic test for cystic echinococcosis based on
Antigen B peptides. Parasite Immunology, 39(12), e12499. https://doi.org/10.1111/
pim.12499.
Petz, L. D. (1993). Drug-induced autoimmune hemolytic anemia. Transfusion Medicine
Reviews, 7, 242–254.
Phillips, E. J., Bigliardi, P., Bircher, A. J., Broyles, A., Chang, Y. S., Chung, W. H., et al.
(2019). Controversies in drug allergy: Testing for delayed reactions. Journal of Allergy
and Clinical Immunology, 143, 66–73.
Piccoli, L., Park, Y. J., Tortorici, M. A., Czudnochowski, N., Walls, A. C., Beltramello, M.,
et al. (2020). Mapping neutralizing and immunodominant sites on the SARS-CoV-2
spike receptor-binding domain by structure-guided high-resolution serology. Cell, 183,
1024–1042.
Pichler, W. J. (2008). The p-i concept: Pharmacological interaction of drugs with immune
receptors. World Allergy Organization, 1, 96–102.
Pichler, W. J. (2019). Immune pathomechanism and classification of drug hypersensitivity.
Allergy, 74, 1457–1471.
Poulter, L. W., Seymour, G. J., Duke, O., Janossy, G., & Panayi, G. (1982). Immunohisto-
logical analysis of delayed-type hypersensitivity in man. Cellular Immunology, 74,
358–369.
Prausnitz, C., & Kustner, H. (1921). Studien aber die Oberempfindlichkeit. Zentralblatt fur €
Bakteriologie: I. Abt. Originale C, 86, 160.
Premkumar, L., Segovia-Chumbez, B., Jadi, R., Martinez, D. R., Raut, R., Markmann, A., et al.
(2020). The receptor binding domain of the viral spike protein is an immunodominant and
highly specific target of antibodies in SARS-CoV-2 patients. Science Immunology, 5(48),
eabc8413. https://doi.org/10.1126/sciimmunol.abc8413.
References 221
Profect, M. (1991). The function of allergy: Immunological defense against toxin. The
Quaterly Review of Biology, 66, 23–62.
Rajan, T. Y. (2003). The Gell–Coombs classification of hypersensitivity reactions:
A re-interpretation. Trends in Immunology, 24, 376–379.
Ramsbottom, K. A., Carr, D. F., Jones, A. R., & Rigden, D. J. (2018). Critical assessment of
approaches for molecular docking to elucidate associations of HLA alleles with adverse
drug reactions. Molecular Immunology, 101, 488–499.
Richeldi, L. (2009). Diagnosing latent tuberculosis infection. Guess who’s coming to dinner?
American Journal of Respiratory and Critical Care Medicine, 1-2, 180. https://doi.org/
10.116/rccm.200904-0562ED.
Rixe, N., & Tavarez, M. M. (2021). Serum sickness. In StatPearls. Treasure Island (FL): Stat-
Pearls Publishing. Available from: https://www.ncbi.nlm.nih.gov/books/NBK538312/.
Romano, A., Atanaskovic-Markovic, M., Barbaud, A., Bircher, A. J., Brockow, K.,
Caubet, J. C., et al. (2020). Towards a more precise diagnosis of hypersensitivity to
beta-lactams—An EAACI position paper. Allergy, 75, 1300–1315.
Romano, A., Blanca, M., Torres, M. J., Bircher, A., Aberer, W., Brockow, K., et al. (2004).
Diagnosis of nonimmediate reactions to beta-lactam antibiotics. Allergy, 59, 1153–1160.
Romano, A., & Caubet, J. C. (2014). Antibiotic allergies in children and adults: From clinical
symptoms to skin testing diagnosis. The Journal of Allergy and Clinical Immunology.
In practice, 2, 3–12.
Rozieres, A., Vocanson, M., Saı̈d, B. B., Nosbaum, A., & Nicolas, J. F. (2009). Role of T cells
in nonimmediate allergic drug reactions. Current Opinion in Allergy and Clinical
Immunology, 9, 305–310.
Sanico, A. M., Bochner, B. S., & Saini, S. S. (2002). Skin testing methods. In D. C.
Adelman, T. B. Casale, & J. Corren (Eds.), Manual of allergy and immunology
(pp. 485–486). Philadelphia: Lippincott, Williams & Wilkins.
Sch€onbeck, S., Chrestel, S., & Hohlfeld, R. (1990). Myasthenia gravis: Prototype of the
antireceptor autoimmune diseases. International Review of Neurobiology, 32, 175–200.
Seibert, F. B. (1934). The isolation and properties of the purified protein derivative of tuber-
culin. American Review of Tuberculosis, 30, 713–720.
Sekine, T., Perez-Potti, A., Rivera-Ballesteros, O., Strálin, K., Gorin, J.-B., Olsson, A., et al.
(2020). Robust T cell immunity in convalescent individuals with asymptomatic or mild
COVID-19. Cell, 183, 158–168.
Shrotri, M., van Schalkwyk, M. C. I., Post, N., Eddy, D., Huntley, C., Leeman, D., et al. (2021).
T cell response to SARS-CoV-2 infection in humans: A systematic review. PLoS One, 16,
e0245532. https://doi.org/10.1371/journal.pone.0245532.
Song, Z., Marzilli, L., Greenlee, B. M., Chen, E. S., Silver, R. F., Askin, F. B., et al. (2005).
Mycobacterial catalase-peroxidase is a tissue antigen and target of the adaptive immune
response in systemic sarcoidosis. Journal of Experimental Medicine, 201, 755–767.
Strachan, D. P. (1989). Hay fever, hygiene, and household size. British Medical Journal, 299,
1259–1260.
Szatkowski, J., & Schwartz, R. A. (2015). Acute generalized exanthematous pustulosis
(AGEP): A review and update. Journal of the American Academy of Dermatology, 73,
843–848.
Takhar, P., Corrigan, C. J., Smurthwaite, L., O’Connor, B. J., Durham, S. R., Lee, T. H., et al.
(2007). Class switch recombination to IgE in the bronchial mucosa of atopic and nonatopic
patients with asthma. Journal of Allergy and Clinical Immunology, 119, 213–218.
Takizawa, F., Tsuji, S., & Nagasawa, S. (1996). Macrophage phagocytosis upon iC3b
deposition on apoptotic cells. FEBS Letters, 397, 269–272.
222 CHAPTER 6 DTH methods and immune response to covid
Hesitancy to get
vaccinated against
COVID-19 and how it
might be overcome
7
Charles S. Pavia∗
a
Department of Biomedical Sciences, NYIT College of Osteopathic Medicine, New York Institute of
Technology, Old Westbury, NY, United States
b
Division of Infectious Diseases, New York Medical College, Valhalla, NY, United States
∗
Corresponding author: e-mail address: [email protected]
One evening, as a young teenager in the 1960s, I watched a televised movie entitled
“The Story of Louis Pasteur”. It impressed me so much that I began to seriously con-
sider pursuing a career in medicine or one of the biological sciences. I subsequently
chose the latter career path and, several years, later I received my Ph.D. degree in
microbiology and immunology. The movie was a classic Hollywood production
originally released for public viewing in 1937 and was a “Best Picture” nominee.
The lead character was the accomplished and well-known actor, Paul Muni, who
won a “Best Actor” award from the motion picture industry for his outstanding per-
formance in his portrayal of Pasteur—considered by many in the medical and scien-
tific community to be one of the founding fathers of modern microbiology and
immunology. He was a chemist by training (as well as being a self-made accom-
plished artist), and he coined the term “microbe”, based on his many discoveries
and drawings of what he observed under the microscope often to the displeasure
of some of his contemporaries, especially the medical establishment. Many of its
members were reluctant initially to go along with some of his experimentally derived
ideas and concepts, based in part because they seemed quite radical at the time given
the limitations of biomedical science 150 + years ago. Despite possibly putting his
career and reputation in jeopardy by occasionally going beyond acceptable medical
ethics when his work involved certain types of human studies, which were less re-
strictive then than they are now, Pasteur persevered working tirelessly and with much
conviction to prove his sceptics wrong, and indeed he showed through careful exper-
imentation that things (minute living organisms) not visible to the naked eye can in-
deed cause disease. By doing so, he debunked the previously held belief, by some of
his counterparts, in the theory of “spontaneous generation”, and is credited with
providing us with the germ theory of disease. Eventually, many of his original scep-
tics started to accept his ground-breaking findings as being true.
While some parts of the movie may have been slight exaggerations and overly
dramatic renditions of actual events of those described in Pasteur’s biography
(Vallery-Radot, 1929), from which the movie got much of its material, it nonetheless
had quite a number of heart-warming and inspiring scenes to go along with scientif-
ically accurate pieces of information. Foremost among Pasteur’s many life-altering
achievements was his development of two diverse vaccines—one for preventing
anthrax in livestock animals, and the other for preventing/treating human rabies
(Fig. 1)—both being dreaded diseases having very high levels of mortality, both then
and now, in the absence of appropriate medical intervention. With these historical
accounts highlighting some of the key events in Pasteur’s career as the background,
it is amazing and at times disturbing to observe how certain aspects of this past
scenario from another era is playing out closely today in the wake of the current
COVID-19 pandemic. In this regard, many people, mostly outside the medical com-
munity, but a significant few within, have refused or remain hesitant to receive any of
the available COVID-19 vaccines. Given the seriousness of COVID-19 with its life-
threatening potential and the worldwide emergence of multiple highly contagious/
virulent mutants, such as the Delta variant, of the etiologic agent SARS-CoV-2,
FIG. 1
This relatively large and impressive portrait of Pasteur, done by Albert Edelfeldt in 1885 and
having the dimensions of 155 cm in height and 127.5 cm in width, shows him observing one of
his rabies “cultures” using rabbit spinal cord material that presumably enabled him to grow
the rabies virus. The original painting hangs majestically in the dining room of Pasteur’s
former home/laboratory located on the campus of the Pasteur Institute in Paris.
Hesitancy to get vaccinated against COVID-19 225
this reluctance to get vaccinated is very difficult to understand and has led to tragic
results. In addition, the vaccine options against COVID-19 are multiple (Table 1) and
involve either the use of a purified mRNA component of the virus or a viral vector
formulation, with both types having excellent safety and efficacy results based on
extensive clinical trials and, as of September 2021, have been approved or are near-
ing full approval from the FDA for use in the United States. In July 2021, as part of a
nationally televised briefing (White House COVID-19 Response Team, 2021),
highly regarded public health officials from the U.S. Centers for Disease Control
and Prevention (CDC) and the National Institutes of Health provided updates on a
new surge nationwide in hospitalizations and deaths due to COVID-19, and have
pointed out correctly that we are now entering into a “pandemic of the unvacci-
nated”. This comment by the director of the CDC is supported by data showing that
in some locations > 99% of the current wave of victims developing serious disease
are among the unvaccinated, leading to the obvious conclusion that these negative
outcomes could have been avoided if these victims had chosen to receive the vaccine
beforehand. This surge in cases has continued and even increased in many locations
in the months that followed.
What are the reasons for such hesitancy/resistance or for some of the obstacles to
get vaccinated? Here are some major examples (summarized in Table 2):
• Let’s take a wait and see attitude—observe how vaccine recipients respond/react
to the vaccine, in terms of making sure that the vaccine is safe and there will not
be any negative outcomes, and/or that it really works;
• Fear of needles or being jabbed and having to endure a possibly painful
injection—this is mostly a psychological or behavioural reaction that may have
been triggered by prior unpleasant experiences when receiving injections for
various other medications or from during a routine blood draw;
• Difficulty in gaining access to vaccine sites by the elderly, the severely disabled
and impoverished people, or in mobile inoculation units being able to safely reach
these home-bound or institutionalized people;
• Exercise our “free will” or constitutional rights—in the United States these
claims are often invoked by referring to the first amendment of the US
Constitution, and falling in line with not having to be compelled to take the
vaccine as a requirement to participate in the activities associated with any
organization, location or function, such as a governmental agency, an academic
institution, transportation venues, sport teams, sport/recreational, entertainment
or indoor dining facilities, places of worship, or an employer;
• Moral beliefs held by members of certain religious groups that are against various
forms of medical intervention; and perhaps the most egregious of them all;
• Believing or being swayed by the many forms of misinformation/disinformation
or conspiracy theories, such as stating that the science behind the vaccine’s
development and success is fake, or not to be trusted, or we are being used as
“guinea pigs”, which is being spread over various Internet social media platforms
and some cable news networks by mostly unscrupulous individuals, some of
whom claim to have expertise on medical matters, but actually don’t.
Table 1 Comparison of the key features of the COVID-19 vaccines that are produced by the four leading manufacturers.
Serious
Vaccine Type of Number of Storage Authorized for EUAb pending adverse
manufacturer vaccine doses method Efficacy use in the U.S.a final approval events
Moderna mRNA 2c
4–10 Co
> 94% Yes Yesd Raree
Pfizer mRNA 2c 80 oC > 95% Yes Yesd Raree
Janssen Viral vector 1c 4–10 oC 66.3% Yes Yes Yesf
Astra-Zeneca Viral vector 2 4–10 oC 63%–84% Yes Yesd Yesf
a
Information that was available as of August 2021.
b
In the U.S., an EUA (Emergency Use Authorization) was originally given for these vaccines for people aged >16, which was subsequently expanded to >12 years of
age.
c
As of October 2021, additional booster injections were highly recommended for these vaccines by the FDA and CDC.
d
In the U.K., an EUA no longer applies for the Pfizer, Moderna and Astra-Zeneca vaccines which now have been granted final approval for use. In the U.S. as of
August 2021, the Pfizer vaccine was given FDA approval, while the other 3 are undergoing further evaluation for full approval by the FDA.
e
Milder and temporary forms of myocarditis and pericarditis have occurred in a small number of young adults with these vaccines.
f
Blood clots have been reported in a small number of female vaccine recipients < 50 years of age with these vaccines.
Hesitancy to get vaccinated against COVID-19 227
It is important to realize that the anti-vaccine sentiment is not limited to the United
States, but it is a global problem (Hornsey, Harris, & Fielding, 2018) and had been
well established as a recurring theme multiple times prior to the onset of the current
pandemic often with devastating consequences. Even certain political leaders and
other professionals of varying degrees of influence have joined the anti-vaccine
bandwagon. This is reminiscent of what has occurred in the pre-COVID days with
certain other infectious diseases (reviewed in Smith, 2017), where “herd immunity”
which can develop when a sufficient number of people survive a natural infection,
has been invoked as the only mechanism whereby the spread of SARS-Cov-2 can be
blocked leading allegedly to its eventual disappearance within the general popula-
tion. Evidence in support of the pervasiveness and effects of these untenable reasons
against the COVID-19 vaccine comes from recently published data put forth by the
Kaiser Foundation (Hamel, Kirzinger, Munana, & Brodie, 2021), and these are as
follows.
In terms of demographics, 27% of the people in the United States remains
vaccine-resistant, saying they probably or definitely would not get a COVID-19 vac-
cine even if it were available for free and deemed safe by scientists and public health
officials. Vaccine hesitancy is highest among those who identify as belonging to a
certain political party (42%), those in the 30–49 age group (36%), and rural residents
(35%), especially those living in certain parts of the Midwest and Southeast sections
of the United States where less than 50% of the population has gotten vaccinated, and
where concurrently there has been an alarming new surge (as of July/August 2021) in
COVID-19 cases requiring hospitalization. In addition, 35% of African American
adults (a group that has had to bear a disproportionate burden of the effects of the
pandemic) say they definitely or probably would not get vaccinated, citing as major
reasons that they don’t trust vaccines in general (47%) or that they are worried they
may get sick with COVID-19 from just the vaccine alone (50%). This situation sug-
gests that messages combatting particular types of misinformation may be especially
important for increasing vaccine confidence among this group. Perhaps most aston-
ishingly, are the data showing that as much as one third of those who say they are
considered to be essential workers and 29% of those who perform services in a health
228 CHAPTER 7 Hesitancy to get vaccinated against COVID-19
care delivery setting will not take the vaccine. The resistance of people to receive
COVID-19 vaccines is even more perplexing given the excellent track record of
other vaccines, some of which have been with us for nearly 100 years as part of
routine preventive care. These have been designed to protect us against smallpox,
tuberculosis (with BCG) and polio, followed, more recently, with the DPT (diphthe-
ria, pertussis, tetanus), MMR (measles, mumps, rubella), and chickenpox vaccines. It
should be noted that, in most jurisdictions, the latter three childhood vaccine
combinations are required before young children can start school at the elementary
level.
So, in light of the foregoing, how would a modern-day Pasteur react to the rel-
atively high level of resistance to get vaccinated against COVID-19, and what might
be a solution to this problem? Given his personality for scientific rigour combined
with the overwhelming evidence showing the vaccine’s safety (with few exceptions)
and efficacy, he probably would be highly disappointed with the overall public re-
sponse to getting vaccinated, especially by those who remain unconvinced on why
the vaccine is so important from a public health perspective, and why there is such
lack of confidence given the many advances made in medicine over the past 100 +
years. He would also be very outspoken and adamant against the sceptics who per-
petuate false or misleading information about the vaccine. If requested (and this
would seem highly likely given his prestige), he would probably be making frequent
public appearances on various news programs similar to what the current wave of
public health medical experts are doing now. He might even have his own podcast
where he could dispense valuable information to the misinformed or uninformed
public and reinforce the importance of getting vaccinated. He would probably find
it amusing and perhaps a bit misguided that incentives, such as monetary and other
rewards, are being offered in certain parts of the United States to try to get people
vaccinated, even though vaccines are being administered free of charge in most,
if not all, locations. To him, it would seem like deja vu in terms of what he experi-
enced with the obstacles that he had to face in dealing with his disbelieving scientific
and medical contemporaries, when he was making his groundbreaking and life-
changing discoveries in the late 1800s, at a time when complex biomedical processes
were still poorly understood, and initially underappreciated.
Another variable that awaits more clarity is whether and/or when patients, who
may have acquired some form of natural immunity after recovering from COVID-19,
should get vaccinated, if at all. This provokes the following question: are they no
longer susceptible, or are less vulnerable, to serious disease, after re-exposure to
SARS-CoV-2, that getting vaccinated would be unnecessary? It is believed, how-
ever, that solid immunity against COVID-19 may gradually wane (Centers for
Disease Control and Prevention, 2021a), and thus booster injections with one of
the available vaccines may still be necessary in order to provide optimal protection
against re-infection. A somewhat related recommendation, also coming from the
CDC, states that, starting in September 2021, another or third booster shot will have
to be given to earlier vaccines to maximize vaccine-induced protection and prolong
its durability, and that these booster shots will be offered for all Americans, who had
Hesitancy to get vaccinated against COVID-19 229
received their last (second) dose or had recovered from a prior infection with
SARS-CoV-2 at least 8 months previously.
Beyond what a contemporary Pasteur might be inclined to do, what else could be
done? Various organizations have made numerous suggestions, along these lines, in-
cluding several promoted and offered by the CDC (Centers for Disease Control and
Prevention, 2021b), and a few more that this author will offer. They include the fol-
lowing services that could be implemented. A large number of healthcare profes-
sionals and qualified scientists (primarily microbiologists and immunologists) are
needed to support COVID-19 vaccination efforts nationwide. It is important they re-
ceive the necessary training to effectively meet the demands of their roles. Training
must be ongoing as new COVID-19 vaccines become available and as vaccine rec-
ommendations evolve and more is learned about the vaccines and how to improve
and maintain the vaccination process. In terms of educating the lay public, they
are essential to ensuring that the American population is vaccinated safely as soon
as possible, based on a true understanding on why this is an important undertaking.
Furthermore, as parents’ most trusted source of information on vaccines, paediatric
healthcare professionals play a critical role in helping parents and guardians under-
stand the importance of COVID-19 vaccination and assuring them that COVID-19
vaccines are safe and effective, and that they are important steps in protecting their
children’s health (both physical and mental). Parents need to be reminded that fully
vaccinated people are less likely to spread the virus that causes COVID-19. Getting
all family members 12 years and older (and when recommended, children less than
12 years of age) vaccinated can protect other family members around you, including
people at increased risk for severe illness from COVID-19. Students also need to be
reminded that, after they are fully vaccinated, they will be able to resume many
activities with family and friends, such as going to parties, weddings, graduation
exercises, and other social gatherings that they have missed due to prior restrictions
that were imposed on everyone at the height of the pandemic.
As part of the education process, students should be contacted and encouraged
to learn more from reliable sources derived from rigorously peer-reviewed articles,
especially those found on medically-based Internet sites, such as PubMed, reputable
blog and social media posts, properly mentored student-driven publications and so-
cial groups. A feedback mechanism should be created in order for students to ask
questions and get a meaningful response quickly about COVID-19 vaccination, such
as by using either e-mail, online video conferences (via Zoom, Skype, Facetime or
any other similar provider), or by phone number. Any student concerns or questions
should be proactively addressed and the spread and harm of misinformation should
be countered by sharing credible and accurate information. Students, as well as other
participants, should be warned about relying on unregulated or non-scientifically
based sources of information that are circulating on the Internet or other similar plat-
forms. Students should be warned about the dangers caused by misinformation
and disinformation, and health literacy should be promoted as a means to be fully
informed in understanding the benefits of being vaccinated as well as the negative
outcomes that could arise if the vaccine is not received.
230 CHAPTER 7 Hesitancy to get vaccinated against COVID-19
Although much of the preceding suggestions pertain mostly to the adolescent age
group, similar interventions should also be considered for those attending colleges
and universities after completing high school, to reinforce and update what students
had learned previously about vaccines. Hopefully, as this younger and now well-
informed generation matures into adulthood, they will be representative of a popu-
lation having much greater acceptance and less resistance to getting vaccinated when
deemed necessary by experts in the health care community for both now and, just as
important, later on, when other future serious outbreaks may arise.
As an additional approach, schools, at both the elementary and high school level,
can also take the initiative by recruiting the suitably trained medical professionals or
scientists to come to their classrooms, when this becomes allowable, and supplement
the curriculum by providing a better understanding of the vaccine process. In order to
accomplish this task more effectively, local health departments, especially those
within the jurisdiction of the schools, should provide the schools with a registry
of trained personnel who would be willing to come to the schools and share their
knowledge and expertise pertaining to the COVID-19 vaccines with the students,
along with the teachers and administrators, as well as answering any questions or
concerns that students may have on this topic, preferably at no additional cost to
the school district.
Another possible program worth considering would be for medical schools to of-
fer a short course on the vaccine process to be attended by elementary and high
school teachers to better educate them on this topic with all of its subtle nuances.
Upon completion of the course, the teachers would receive a certificate of recogni-
tion (similar to CME credits) showing that they had participated successfully in this
learning exercise. As such, they would be well equipped to return to their respective
schools with what they had learned about vaccines and share this information with
their students, during one of their standard classroom sessions or remotely (typically
when the subject of “Biology” is being taught or during “Health Class”). Presumably,
the best time to give this course would be during the summertime when most medical
schools are less active with their didactic responsibilities as part of the regular pre-
clinical curriculum, and where most of the first- and second-year medical students
are not on campus due to being on an extended break for their summer vacation.
In so doing, this would not impose an undue burden on the school’s infrastructure
or their faculty who would be providing this pertinent information to the participants
in this program.
In conclusion, people should not be fearful of receiving any of the available anti-
COVID-19 vaccines. The benefits of getting jabbed far outweigh the minimal risks
of having an adverse or serious outcome. Historically, vaccines have had an excellent
track record in terms of saving lives, reducing morbidity caused by a wide variety of
infectious agents, and easing the burden of the health care community and infrastruc-
ture. In addition, the various and somewhat innovative interventions put forth in this
chapter, that are designed to educate people about the COVID-19 vaccines, with the
goal of getting as many people vaccinated, especially towards dealing with the exist-
ing anti-vaccine trend, may be seen as a daunting task. Accordingly, it will require a
References 231
References
Centers for Disease Control and Prevention. (2021a). Frequently asked questions about
COVID-19 vaccination. 19 August 2021. https://www.cdc.gov/coronavirus/2019-ncov/
vaccines/faq.html#::text¼Yes%2C%20you%20should%20be%20vaccinated,ve%20al
ready%20had%20COVID%2D19.
Centers for Disease Control and Prevention. (2021b). How schools can support COVID-19
vaccination. June 29, 2021. https://www.cdc.gov/vaccines/covid-19/planning/school-
located-clinics/how-schools-can-support.html.
Hamel, L., Kirzinger, A., Munana, C., & Brodie, M. (2021). KFF COVID-19 monitor:
December 2020. https://www.kff.org/coronavirus-covid-19/report/kff-covid-19-vaccine-
monitor-december-2020/. accessed on July 22, 2021.
Hornsey, M. J., Harris, E. A., & Fielding, K. S. (2018). The psychological roots of anti-
vaccination attitudes: A 24-nation investigation. Health Psychology, 37(4), 307–315.
https://doi.org/10.1037/hea0000586.
Pasteur, L. (1939). Discours prononce à Douai, le 7 decembre 1854, à l’occasion de l’instal-
lation solennelle de la Faculte des lettres de Douai et de la Faculte des sciences de Lille. In
P. Vallery-Radot (Ed.), Vol. 7. Speech delivered at Douai on December 7, 1854 on the
occasion of his formal inauguration to the Faculty of Letters of Douai and the Faculty
of Sciences of Lille (p. 131). Oeuvres de Pasteur; Paris, France: Masson and Co. http://
gallica.bnf.fr/ar k:/12148/bpt6k7363q/f137.chemindefer.
Smith, T. C. (2017). Vaccine rejection and hesitancy: A review and call to action. Open Forum
Infectious Diseases, 4(3). https://doi.org/10.1093/ofid/ofx146.
Vallery-Radot, R. (1929). The life of Pasteur. Garden City, NY: Garden City Publishing
Co., Inc.
White House COVID-19 Response Team. (2021). White House COVID-19 Response
Team Briefing. https://www.c-span.org/video/?513463-1/cdc-director-warns-pandemic-
unvaccinated. July 16, 2021.
This page intentionally left blank
CHAPTER
The emergence of
SARS-CoV-2 variants
of concern in Australia
by haplotype coalescence
8
reveals a continental link
to COVID-19 seasonality
Tre Tomaszewskia, Volker Gurtlerb, Kelsey Caetano-Anollesc, and Gustavo
Caetano-Anollesa,∗
a
Evolutionary Bioinformatics Laboratory, Department of Crop Sciences, University of Illinois,
Urbana, IL, United States
b
RMIT University, Melbourne, VIC, Australia
c
Callout Biotech, Albuquerque, NM, United States
∗
Corresponding author: e-mail address: [email protected]
1 Introduction
The COVID-19 pandemic illustrates how a virus is capable of overcoming barriers to
its persistence by rapidly changing its genomic makeup. Thanks to extensive world-
wide genome sequencing efforts, researchers now have direct access to information
about the levels of genetic variation unfolding in the evolving viral population, as
well as variations associated with physiological responses of human or animal hosts.
As of January 15, 2022, the GISAID initiative (https://www.gisaid.org) sponsored by
many governments in partnership with public health and research institutions
(Elbe & Buckland-Merrett, 2017; Khare et al., 2021; Shu & McCauley, 2017) has
collected over 7 million genomic sequences of the SARS-CoV-2 virus, making them
freely accessible to the scientific community for analysis. In parallel, the open-
source Nextstrain project (https://nextstrain.org) made available a continuously
updated phylogenomic view of this data alongside with powerful and portable
analysis and visualization tools. These resources provide a unique window into
our evolutionary understanding of a human pathogen of great significance.
Genetic variation refers to the existence of differences among the genomes of a
set of closely or more distantly related organisms or viruses. This diversity in
Methods in Microbiology, Volume 50, ISSN 0580-9517, https://doi.org/10.1016/bs.mim.2022.03.003
Copyright © 2022 Elsevier Ltd. All rights reserved.
233
234 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
genomic makeup constitutes one primary source of phenotypic diversity, i.e., diversity
in observable biological characteristics (traits). The other primary source involves epi-
genetic variation. Genetic variation results from the effects of a multiplicity of pro-
cesses, including spontaneous mutation, error-prone replication, recombination, and
genetic exchange. Mutations can be small-scale or large-scale alterations in the nucle-
otide sequence of genomic DNA present in most life forms or RNA typical of some
viruses. Small-scale alterations include exchange (substitution), addition (insertion) or
removal (deletion) of nucleotides in a sequence. Large-scale alterations include dupli-
cations, translocations or inversions of larger nucleic acid segments. Regardless of
their nature, the physiological impact of these alterations typically materializes at
the level of proteins or functional RNA. Nucleotide triplets for the most part encode
for amino acids, which control the physiological activities of the cell by serving as the
building blocks of proteins. A non-synonymous mutation leading to change in one or
more amino acids of a polypeptide sequence can alter the structure and functioning of
the mutated protein. For example, in the case of the SARS-CoV-2 virus, a single amino
acid substitution at position 614 of the viral spike glycoprotein (from aspartic acid to
glycine, referred to as the D614G mutation) that occurred during the first wave of the
pandemic resulted in increased viral transmissibility (Voltz et al., 2021). Distinct
viruses holding one or a unique constellation of these types of mutations are generally
called “variants” (Lauring & Hodcroft, 2021). At the protein level, mutations cause
amino acid substitutions (e.g., D614G), which are called “amino acid variants.”
Genetic variants arise in the context of evolving populations. Thus, mutations in
single or multiple genomic locations are often the subject of evolutionary effects
on fitness (e.g., natural selection) or the effect of chance events on sampling
(e.g., genetic drift). In the case of viruses, their fate can depend for example on
whether they confer competitive advantage to viral replication, rates of transmission,
immune escape, or virulence. Mutations that do not provide an advantage are often
eliminated from the population, unless “founder effects” on newly established viral
populations extend their persistence. Epidemiologically, a mutation that alters trans-
missibility, disease severity, or immune or vaccine escape becomes a “mutation of
concern” (MOC) and its presence in a variant a candidate for surveillance and re-
sponse. More importantly, a “variant of concern” (VOC) is a variant of the virus
exhibiting a constellation of mutations associated with statistically significant and
experimentally verified increases in virus transmissibility, disease severity, immune
and vaccine escape, diagnostic test evasion, or other clinical or epidemiological
criteria of significance. VOCs become immediate priority for surveillance and
response, especially when their prevalence increases worldwide.
Haplotypes are sets of mutations that are often inherited together. In the case of
viruses, haplotypes are known to represent mutations that appear tightly linked with
each other. For example, the D614G mutation of the SARS-CoV-2 spike protein is
part of an haplotype of four mutations that also alter the NSP12 polymerase (P323L),
50 untranslated region (UTR), and silently the NSP3 papain-like protein (F106F).
This haplotype was the first gene set to be fixed in the worldwide viral population
during the first wave of the COVID-19 pandemic in early 2020. Since VOCs are
2 Methods 235
mutation constellations reflecting successful viral variants that have overtaken the
global population, there is an implicit assumption that these constellations are stable
haplotypes. This assumption however has not been fully tested. Here we explore the
appearance and accumulation of major mutations typical of VOCs in Australia as
the viral disease progresses towards becoming endemic. We study the constellation
of mutations characteristic of VOCs to determine if the mutation sets acted as hap-
lotypes and to test if these haplotypes are the subject of regional variation in
Australia. Our goal is to explore processes behind the emergence of VOCs in a viral
pandemic, including effects of viral seasonal behavior.
2 Methods
The metadata for 7,175,152 SARS-CoV-2 genome sequences was downloaded from
GISAID (https://www.gisaid.org) on January 18, 2022. The metadata were then
filtered for sequences marked “complete” with “human” hosts and a “location” field
containing the case-insensitive term “Australia,” which reduced the set to 58,378
sequences. Of these, the sequences were collected between January 1, 2020 and
January 13, 2022 (743 days) and submitted for deposition to GISAID between
January 31, 2020 and January 17, 2022 (717 days) (see acknowledgements in
Supplementary information in the online version at https://doi.org/10.1016/bs.
mim.2022.03.003 for complete list of Accession IDs used).
The Australian region (state/territory) for each sequence was then extracted from
the “location” field, resulting in sequences belonging to each of eight regions
(Table 1). The metadata was then labeled by “period,” which was derived from
the collection date’s year and calendar quarter. This was done so that, for example,
January 1, 2020 to March 31, 2020 was designated as “Period 1” and January 1, 2022
to March 31, 2022 as “Period 9”.
The sequence metadata provide the field “AA Substitutions,” which contains a
comma-separated list of each identified amino acid substitution (against the refer-
ence sequence NC_045512) by protein name, reference amino acid, amino acid
location within the sequence, and the substituting amino acid (formatted as < Protein
Name >_<Reference AA ><AA Protein Location ><Substitution AA >). The list
for each sequence was transformed into a one-hot encoding for each of the 9281 mu-
tational substitutions, indexed by the Accession ID, and the derived region and
period.
Grouped by these derived attributes, a simple summation of each possible muta-
tion across sequences provided the occurrence count for each region-period group-
ing. Dividing the summation of any mutation by the total number of sequences within
the group provided the prevalence of each amino acid substitution for each region-
period. These groups were then aggregated and regrouped by mutation, enabling
regional comparisons of the prevalence of each mutation by time period (year-
quarter).
Since substitution groups containing low variance or spurious substitutions were
undesired for further analysis, the groups were filtered by a “relevancy” heuristic.
The prevalence of any given substitution was required to be above a threshold of
0.1 in more than 2 regions for at least one period, although there was no requirement
that this threshold was met during the same period.
Review of the initial results revealed certain variant-specific substitutions occur-
ring in improbable time-periods (e.g., simultaneous mutations appearing in signifi-
cant amounts 3 quarters prior to announced detection). Further analysis of the data
indicated 292 instances were labeled with collection dates that were only identified
by year. These were reconciled by using the submission date as a proxy. The entire
process detailed above was then repeated to achieve final results.
Extraction and transformation was performed using the Python library Pandas
(McKinney, 2010; Pandas Development Team, 2022). The Python library
“matplotlib” (Caswell, Droettboom, Lee, et al., 2021; Hunter, 2007) was used to
produce raw plots of the data, followed by an additional arrangement, annotation,
and graphical modification using Adobe Illustrator. Source code and supplementary
information can be found at https://doi.org/10.1016/bs.mim.2022.03.003.
3 VOCs in Australia
SARS-CoV-2 variants are organized around a master genomic sequence of the virus
that originated in the city of Wuhan in China (accession NC_045512.2, version
March 30, 2020; previously “Wuhan seafood market pneumonia virus”). Many
mutations have been added to, and subtracted from, this master sequence since
the beginning of the pandemic. These genomic changes can be traced through their
phylogenies (Fig. 1A). Phylogenies are hypotheses of history and genealogical
relationship among groups of genomes (evolving taxa) in the form of tree structures
(networks without reticulations). They harbor specific connotations of ancestry
and an implied time axis, which enables the study of important epidemiological
phenomena such as viral spread, variant introduction, and rates of genomic
change and epidemic growth. Splits in the branches of these trees define clades,
FIG. 1
The mutational landscape of the SARS-CoV-2 virus at the beginning of 2022 and its historical
spread throughout the Australian continent. (A). A maximum likelihood phylogenetic tree
describes the worldwide history of the SARS-CoV-2 genome. The timetree of 3347 genomes
randomly sampled between December 2019 and January 2022 was obtained from Nextstrain
(https://nextstrain.org) on January 15, 2022. The tree unfolds time of genome collection date
from left to right. Its leaves (taxa indicated with circles) are colored according to clade (group
of taxa with a common evolutionary origin) and emerging variants of concern (VOCs)
nomenclature. The origin of VOCs occurs when a clade originates along branches of the
phylogeny. Note the early arrival of VOC alpha, followed by VOC delta and then VOC omicron.
The timeline of clades and VOCs show three successive phases driven by proteome flexibility
and rigidity, environmental sensing and vaccine-driven immune escape, which are shaded in
light yellow, blue and salmon, respectively (Caetano-Anolles, Hernandez, Mughal,
Tomaszewski, & Caetano-Anolles, 2022). (B). Plots show numbers of newly confirmed cases
per 1000 people (in logarithmic scale and as 7-day rolling averages) and smooth percentages
of genomes holding major VOCs in Australia since the beginning of the recorded COVID-19
pandemic. COVID-19 and genome data are derived from Johns Hopkins Univ., CSSE and
GISAID, respectively. (C). Spike map showing the population density of Australia as a grid of
vertical bars depicting number of people per square kilometer of land area (courtesy of
Alasdair Rae, Automatic Knowledge Ltd., Sheffield, UK). The different states/territories of
Australia are identified with colored numbers using shades that correspond to increasing
latitudes of their population medians across cells (from red to turquoise). The pie chart
describes the relative number of total cases (cumulative, confirmed and under investigation)
reported by the Department of Health, States and Territories for individual regions on
February 4, 2022.
238 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
i.e., groups of taxa with a common evolutionary origin. Clades are often defined by
the statistical distribution of distances between phylogenetic clusters followed by
lineage merging based on mutations that are shared. As of February 2022, genome
sequences have been clustered into 11 GISAID clades (L, S, O, V, G, GH, GR, GV,
GRY, GK, and GRA) or 23 Nextstrain clades defined by a year-letter nomenclature.
In the case of Nextrain clades, a new clade must differ by at least 2 mutations from its
parent major clade (Hodcroft, Hadfield, Neher, & Bedford, 2020). The tree shown in
Fig. 1A uses the Nextstrain nomenclature to pinpoint the evolutionary appearance of
major VOCs along a timeline that originated when the first two clades (19A and 19B)
diverged from each other. In the figure, the current VOC omicron wave is repre-
sented by major clades 21 K and 21 L, which originated from a larger more basal
clade that gave rise to VOC alpha. Nextstrain clades of VOC omicron correspond
to the recent GISAID clade GRA. In addition, clades can be defined at lower gran-
ularity using the Phylogenetic Assignment of Named Global Outbreak LINeages
(Pangolin) tool that automatically assigns sequences to lineages and sublineages
(Rambault et al., 2020). For example, VOC omicron corresponds to Pangolin lineage
B.1.1.529 and the previously prevalent VOC delta to lineage B.1.617.2, both of
which harbor numerous sublineages.
VOCs emerged in October 2020, less than half a year after the first wave of the
pandemic. VOC alpha (also known as Nextstrain clade 20I or Pangolin lineage
B.1.1.7) appeared in the United Kingdom and was the first to expand quickly world-
wide, probably correlated with significant increases in transmissibility and infection
rates (Davies et al., 2021). VOC beta (20H, B.1.351) appeared in December 2020,
following its first report in South Africa, and VOC gamma (20 J, P.1) appeared in
the Amazonian region of Brazil in January 2021. The highly prevalent VOC delta
(21A, B.1.617.2), while first discovered in India in October 2020, became predominant
worldwide in June 2021, almost completely replacing other developing VOCs. Finally,
VOC omicron was first identified in Botswana and South Africa early in November
2021 and is currently sweeping the world, replacing VOC delta. A global analysis
of the spread of the different VOCs (except omicron) and an estimate of effective
reproduction numbers revealed rapid replacement of previously circulating variants
and transmissibility increases ranging from 25% (alpha) to 97% (delta) (Campbell
et al., 2021). These estimates are expected to increase substantially with VOC omicron.
We here focus on the COVID-19 pandemic in Australia and the effects that lat-
itude has on the establishment of VOC-induced disease. Compared to responses from
the US and European countries, the disease mitigation strategies employed by fed-
eration and local governments of the Australian Commonwealth have been swift and
effective. This provides a unique opportunity to study VOC emergence at many lat-
itude levels in a country that has been able to control infection for the majority of the
pandemic (Fig. 1B). The first confirmed case of COVID-19 was identified in Vic-
toria on January 25, 2020. Both the central government and individual states
responded swiftly to the outbreak by closing borders. This controlled the first wave
to some degree by the beginning of April. However, a second and more deadly wave
emerged in Victoria during May and June 2020. Although it was largely localized to
3 VOCs in Australia 239
Melbourne, it was considerably more widespread than the initial wave. Strict lock-
down managed to control the disease by November 2020. In order to curb cluster
outbreaks, Australia pursued a zero-COVID public health policy of suppression
(i.e., “find, test, trace, isolate and support”) that minimized domestic community
transmission, enforced strict international border controls, and curbed local out-
breaks via lockdowns and exhaustive contact tracing. The policy lasted until late
2021. Despite efforts and a nationwide vaccination program, VOC delta levels in-
creased in April 2021 and a “delta wave” overtook the country in June 2021 with
a significant outbreak in New South Wales. Major city lockdowns during July
through December 2021 were unable to suppress the rise of case numbers, which
was notably exacerbated by the VOC omicron wave that began in 2022. At the be-
ginning of December 2021 there were 211,654 reported cases. After only 2 months
(up to February 4, 2022), that number of total cases increased to 2,319,029, with
940,596 cases corresponding to New South Wales and 870,416 to Victoria. Despite
these large case numbers, only 4073 total deaths were reported for the entire country.
We note that the “delta wave” that started in April 2021 and was predominant for a
period of 5–6 months was largely responsible for a significant number of these
deaths. The first cases of VOC omicron were reported in Sydney on November
28, 2021, and in Darwin and Sydney on November 29, 2021, all infected travelers
returning from southern Africa. Fig. 1B describes the percentage of sequenced ge-
nomes corresponding to the main VOCs alpha, delta and omicron that were present
in Australia. Remarkably, VOC omicron (21K) took over the identity of most of ge-
nome samplings in less than a month, replacing the fully prevalent VOC delta (21J).
Australia is inhabited by 26 million people, making the country the most popu-
lous in Oceania. However, because of its significant size (the 6th largest nation in the
world), Australia has a very low population density of 3 people/km2. Furthermore,
most people live in major urban areas, which largely correspond to the capital cities
of the state/territories. The largest cities include Sydney (4.6 million in habitants),
Melbourne (4.2 million), Brisbane (2.2 million), Perth (1.9 million) and Ade-
laide (1.2 million). The Gold Coast, Newcastle and Wollongong add an extra 1.2
million. Fig. 1C shows a spike map of the population density of Australia. It iden-
tifies the different states/territories of Australia with numbers colored according to
the latitudes of their population medians: 1, Northern Territory (12°S); 2, Queens-
land (27°S); 3, Western Australia (32°S); 4, New South Wales (33°S); 5, South
Australia (35°S); 6, Australian Capital Territory (ACT) (35.2°S); 7, Victoria
(39°S); and 8, Tasmania (43°S). Regions 1–4 can be dissected from regions
5–8 by a –34°S latitude transect, separating the largest cities of Sydney and Mel-
bourne from each other by 4°, 713 km air distance, and a state boundary half-way
between the two cities. Most reported COVID-19 cases correspond to the largest cit-
ies located within a 30°S to 50°S latitude corridor, which was previously iden-
tified to be associated with seasonality during the first wave of the pandemic
(Caetano-Anolles et al., 2022). A pie chart describing the proportion of total cumu-
lative cases in states/territories shows that 88% of cases appeared in New South
Wales and Victoria, driven mainly by the Sydney and Melbourne metropolitan areas
240 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
report in Germany and spread throughout continents during the January–April period
of 2020. The haplotype is the most stable so far and is believed to be linked to
increases of COVID-19 infectivity (Becerra-Flores & Cardozo, 2020; Korber
et al., 2020). Mutations were already present in most Australian regions at 60% prev-
alence levels during the first quarter of 2020, but at lower levels in Tasmania, ACT
and Southern Australia. Remarkably, while these markers of haplotype 5 were max-
imally prevalent worldwide, their prevalence only reached 100% in the third quarter
of 2021 for all regions of Australia. We also note that the emergence of the haplotype
was noisy and sometimes decoupled across regions until the second quarter of 2021.
Temporal decoupling was evident in the Northern Territory, Queensland, New South
Wales, and Western Australia, i.e., regions above the 34°S latitude transect that are
warmer. The slow establishment of haplotype 5 may be explained by the zero-
COVID public health policy of the country.
The R203K and G204R markers of the nucleocapsid protein (N-protein) emerged
as a tightly linked haplotype (haplotype 2) in all regions during the start of the pan-
demic (Fig. 3B). Mutation decoupling occurred between the third quarters of 2020
and 2021 in Queensland and Victoria. Prevalence reached highest levels between the
third quarter of 2020 and the first quarter of 2021 in regions above the 34°S latitude
transect that are colder, especially in ACT, Victoria and Tasmania (which reached
100% prevalence levels), but was found to significantly decrease in regions closer
to the Equator. These patterns and the rise of the haplotype during the winter in
the Southern Hemisphere suggest a seasonal effect. We note that the haplotype dis-
appeared from Australia in the third quarter of 2021 when VOC delta took over the
viral population but later re-emerged forcefully as a tightly linked haplotype with the
rise of VOC omicron at the end of 2021. These mutations are located in the serine/
arginine-rich linker that separates the N-terminal and C-terminal RNA-binding do-
mains of the N-protein, which we found is intrinsically disordered (Tomaszewski
et al., 2020).
FIG. 4
Core haplotypes of VOC alpha, delta and omicron. (A). Accumulation plots describing the
prevalence of the 12 amino acid variants of haplotype 1 of VOC alpha. The plot overlap
describes the tight curve overlap of the 12 markers, revealing only a decoupling pattern in the
curves of variant R52I of the ORF8 protein. (B). Accumulation plots describing the prevalence
of the 8 amino acid variants of haplotype 6 of VOC delta. The “plot overlap” describes the tight
curve overlap of the 7 markers. Note that there was decoupling associated with variants
P1469S of NSP3 and T77A of NSP6. (C) Accumulation plots describing the prevalence of
representative variants (first in the lists) of the 6 haplotypes that make up the core of VOC
omicron (haplotypes 12–17). For all panels, plots describe variant accumulation in the 8
regions of Australia and are labeled with the name of the amino acid variant colored according
to its presence in the mutant constellation reported for VOCs worldwide (green, VOC alpha;
blue, VOC delta; red, VOC omicron; black, other). The icons for the alpha, delta and omicron
haplotype cores are being used in the network of Fig. 5.
haplotype across regions was low (below 60%) since it follows the low prevalence
of VOC alpha (see Fig. 1B). However, the haplotype was surprisingly absent in
Tasmania.
The core mutant constellation of VOC delta involved an 8-variant haplotype
(haplotype 6) affecting 6 proteins—the N-protein, ORF7b, the NSP3 protease,
4 Prevalence of amino acid variants in Australia 245
NSP4, NSP6 and the NSP14 exonuclease (Fig. 4B). Minor differences in variant
accumulation were observed in P1469S of NSP3 (most notably in Victoria and the
Northern Territory) and in T77A of NSP6 (in South Australia and the Northern
Territory). Remarkably, haplotype prevalence reached 90–100% in regions below
the 34°S latitude transect (South Australia, ACT, Victoria and Tasmania) during
the third quarter of 2021, while for example, it reached only 60% in New South Wales.
Finally, the core mutant constellation of VOC omicron involved a set of 6
haplotypes (haplotypes 12–17) containing 2–8 variants, each of which affected
1–5 proteins, including the S-protein, N-protein, and the membrane (M) and enve-
lope (E) structural proteins, NSP3, NSP5, NSP6 and NSP14 (Fig. 4C). Haplotypes
13, 16 and 17 affected sites exclusively present in the S-protein. Haplotypes 12 and
14 also involved a significant number of S-protein markers. Accumulation curves
showcase how VOC omicron overtook the entire viral population in all regions
and in a period of only two calendar quarters, the last quarter of 2021 and the first
quarter of 2022. Curve overlaps for variants in each haplotype revealed an absence of
significant decoupling patterns. We note, however, that all six haplotypes harbored
common patterns of accumulation, which suggests they exhibit similar behavior
across regions. This merits placing these haplotypes within a single haplotype core.
We also note that S-protein variants A67V and V143del of haplotype 14 and N210del
and N212I of haplotype 17 appeared in advance of VOC delta in the first quarter of
2021, which suggests that these markers were recruited from markers that were al-
ready present in the viral population in early 2021. Furthermore, the N-protein var-
iant P13L of haplotype 12, which is associated with the N-terminal region of the
nucleocapsid that is intrinsically disordered (Tomaszewski et al., 2020), appeared
during the first wave of the pandemic in the first quarter of 2020, reaching very high
prevalence levels in Tasmania. This marker, which was part of a predicted pathway
of mutational change involving protein flexibility/rigidity (Tomaszewski et al.,
2020), likely represents the oldest variant of the VOC omicron haplotype core.
and omicron, i.e., variants S96I, G142D and T478K of the S-protein and variant
T492I of NSP4. Only the most ancestral haplotype, haplotype 5 mentioned above,
unified all three VOCs.
4.4 Amino acid variants that are not part of established VOC
constellations
We identified a number of amino acid variants with significant prevalence that did
not belong to established VOC constellations (see CoVariants; https://covariants.
org). Accumulation plots are shown in Fig. S4 in Supplementary information in
the online version at https://doi.org/10.1016/bs.mim.2022.03.003. Mutants E484K
and A701V of the S-protein, T205I of the N-protein, T85I of NSP2, K835N of
NSP3, and K90R of NSP5 followed increases that mirrored those of VOC alpha
markers. Similarly, G215C of the N-protein and V71I of ORF7a followed increases
similar to those of VOC delta. Finally, V1069 of NSP3 increased together with the
emergence of VOC omicron. Mutants with increases that mirrored those of VOC al-
pha markers reached 100% prevalence in ACT during the last quarter of 2020. An-
other set reached prevalence in ACT during the second quarter of 2021 (R385K of
the N-protein, P129L of NSP2, H1274Y of NSP3, and H234Y of NSP15). We cannot
explain why the ACT region fostered all of these mutations at high levels. The high
prevalence reached by S197L of the N-protein and F308Y of NSP4 in Tasmania can-
not be explained either, especially because both mutations had patterns of accumu-
lation that were linked (suggesting an haplotype). They may represent specific
mutational bursts that occurred in those regions.
FIG. 5
A network of haplotypes describes the emergence of major VOCs in Australia. Nodes are
either haplotypes or individual amino acid variants coalescing into VOC-specific
constellations. Edges describe common patterns of prevalence in accumulation plots. Circles
portray levels of haplotype coalescence. Outer-most circles host amino acid variants and
haplotypes shared between VOCs. Haplotypes are labeled with numbers and variants are
labeled with names that follow accepted nomenclature from the Human Genome Variation
Society. Names are colored according to their presence in established VOCs worldwide or in
black when uniquely present in Australia.
N-protein and NSP6, respectively. In turn, VOC omicron shares only 4 variants with
VOC delta. These markers, 3 of which are S-protein variants, coalesce into VOC
delta’s core. In other words, VOC alpha contributed almost half of its markers
(11 out of 24 total) and 4 out of its 5 haplotypes to VOC omicron, while VOC delta
contributed only 17% of its markers (6 out of 35 total) and only 1 out of its 7
haplotypes to the makeup of the new VOC.
Going back in time, a number of omicron-specific variants were already present
in significant number during the first wave of the pandemic early in 2020.
248 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
These include the S477N variant of the S-protein and the P13L variant of the
N-protein. Other omicron-specific markers appeared by the end of 2020 and begin-
ning 2021 in Australia, including K417N and to a lesser level A67V, N440K, H655Y,
N679K, D796Y of the S-protein and K38R, S1265del, and L1266I of NSP3 (Fig. S3
in Supplementary information in the online version at https://doi.org/10.1016/bs.
mim.2022.03.003). Similarly, several delta-specific variants were already present
in 2020 and very early in 2021, including A222V, L452R and P681R of the
S-protein, 182 T of the M-protein, D377Y of the N-protein, P822L of NSP3,
A446V of NSP6, T181I of NSP6 and to a lesser level V82A of ORF7a, T40I of
ORF7b, P822L of NSP3, P1228L of NSP3, and P77L of NSP13 (Fig. S2 in Supple-
mentary information in the online version at https://doi.org/10.1016/bs.mim.2022.
03.003). Finally, alpha-specific variants were already significantly present during
the end of the first wave, including R52I of ORF8 and T183I of NSP3 (Fig. S1 in
Supplementary information in the online version at https://doi.org/10.1016/bs.
mim.2022.03.003). Many of these reused markers were part of several haplotypes.
In order to strengthen the argument of significant reuse of markers in variant
combinations, we explored their presence in 137,605 sequences of the S-protein re-
trieved worldwide on November 14, 2020 (Showers, Leach, Kechris, & Strong,
2022). For VOC omicron, D614G of haplotype 5, H69del, V70del and Y144del
of haplotype 3 (shared with VOC alpha), D796Y and N679K of haplotype 12,
A67V, Y143del and T547K of haplotype 14, and N440K of haplotype 19, were pre-
sent in the dataset sometimes in combination with others. For example, H69del and
V70del variants of haplotype 3 were present in 1066 genomes as a H69del-V70del-
N439K-D614G combination and in numerous other arrangements in another 698 se-
quences, including 22 sequences of a 10-variant combination of 6 VOC omicron-
specific and 3 VOC-alpha-specific markers (H69del-V70del-Y145del-N501Y-
A570D-D614G-P681H-T716I-S982A-D1118H). Thus, 5 out of 8 haplotypes affect-
ing S-proteins in VOC omicron recruited markers appearing in 2020. Other free-
standing markers were also recruited, including S477N, D574Y, G339D, G142D
and T478K (both shared with VOC delta), and P681R (shared with VOC alpha).
In particular, S477N was present in 8080 sequences as the second most popular com-
bination of the set (S477N-D614G). For VOC delta, T19R and D950N of haplotype 7
(the only S-protein markers in haplotypes), G142D and T478K shared with VOC om-
icron, P681R, L452R and A222V were also present in the dataset. In particular,
A222V appeared in 7088 sequences as a L18F-A222V-D614G combination and
in 169 sequences as a L5F-A222V-D574Y-D614G-H655Y combination (which in-
cludes H655Y of VOC omicron). As expected for VOC alpha, we found 22 instances
of the 10-variant combination containing all S-protein markers of haplotype 1
(A570D, T716I, S982A, D1118H), haplotype 3 (H69del, V70del, Y145del) and hap-
lotype 5 (D614G) and the two free-standing variants N501Y and P681H that collec-
tively characterize the S-protein constellation of this viral variant (H69del-V70del-
Y145del-N501Y-A570D-D614G-P681H-T716I-S982A-D1118H). Recall that the
first appearance of VOC alpha occurred a few weeks before the sampling date of
the dataset in the United Kingdom. To summarize, 16 S-protein variants of VOC
omicron, 7 of VOC delta and all variants of VOC alpha were already present before
5 A network view of haplotype diversity and VOC emergence 249
FIG. 6
A network of SARS-CoV-2 proteins mediated by haplotypes. Nodes are proteins and lines of
the graph are protein interaction expressed as joint protein presence in an haplotype. Pie
charts are nodes describing the relative number of haplotypes made up of only variants of that
protein in the mutant VOC constellations. Pie slices and lines of the graph corresponding to
VOC alpha, delta and omicron are colored green, blue and red, respectively.
the haplotypes of VOC delta but was later (and forcefully) regained in VOC omicron
through S-protein links to both N-protein (haplotype 12) and NSP3 (haplotype 14),
N-protein-specific haplotype 18, NSP3-specific haplotype 20, and 6 core haplotypes
with a multiplicity of S-protein markers. This solidification of the functionalities of
the spike, nucleocapsid and NSP3 papain protease in VOC omicron makes evident
their well-known centrality in viral transmissibility, disease severity, and immune
escape. The S-protein plays critical roles in viral attachment to host cells. Its highly
immunogenic properties and roles in transmissibility and virulence has made the
spike glycoprotein trimer a target for drug and vaccine mitigation (Harvey et al.,
2021). The N-protein, which packages the RNA genomes, is the most abundant viral
protein and is essential for replication, virion assembly, and regulation of the viral
life cycle (Bai, Cao, Liu, & Li, 2021). In addition, the two structural domains of
the N-protein are separated by an intrinsically disordered linker that is highly sensi-
tive to proteolysis and generates at least five proteoforms that bind structured RNA
(Lutomski, El-Baba, Bolla, & Robinson, 2021). This endows the N-protein with a
host of regulatory and immunogenic properties. Finally, the multidomain NSP3
papain-like protease acts on the viral polyproteins, interacts with other NSPs and
RNA to form the replication/transcription complex, antagonizes the host innate
immune response, and supports viral survival (Lei, Kusov, & Hilgenfeld, 2018).
The central role of these three proteins is enhanced in VOCs by an additional central
hub associated with authophagy, the NSP6 protein (Fig. 6). NSP6 has been shown to
induce autophagosome formation and NLRP3 inflammasomes, mediating caspase-1
activation and secretion of pro-inflammatory cytokines known to induce
5 A network view of haplotype diversity and VOC emergence 251
inflammatory cell death (Cottam et al., 2011; Sun, Huang, Xu, & Hu, 2021). The
inflammasomes are multimeric sensor proteins that are critical components of the
innate immune system. However, their aberrant activation can cause serious disor-
ders, including cascades leading to the severe acute respiratory syndrome (SARS)
caused for example by SARS-CoV-2 (Rodrigues et al., 2021). The NSP6-specific
haplotype 4 of VOC alpha shows an early central role in this aberrant activation
of the innate immune system, which was later complemented by the delta-specific
core haplotype 6 and haplotype 9, which are linked to N-protein and ORF3a. Re-
markably, the ORF3a viroporin has been shown to inhibit autophagosome-lysosome
fusion by interacting with a protein of the homotypic fusion and protein sorting
(HOPS) complex (Zhang et al., 2021). This mechanism helps the virus escape deg-
radation. The central role of NSP6 continues in VOC omicron with markers of hap-
lotype 4, haplotype 15 and an extra free-standing NSP6 marker (Figs. 5 and 6), which
prompts evaluation of how VOC omicron mutations are softening aberrant immunity
activations.
The rise of VOC delta haplotypes appear to optimize interactions of the N-protein
with the M-protein and S-protein, and VOC omicron haplotypes similarly now
optimize interactions between the M-protein and both the E-protein and the
S-protein (Fig. 6). Intraviral interactions between these three structural proteins have
been reported to play essential roles in the viral life cycle (e.g., Artika, Dewantari, &
Wiyatno, 2020). They hijack the cellular network of the host. The central intrinsi-
cally disordered serine/arginine-rich spacer of the N-protein that separates its two
domains is vital for effective viral replication. The transmembrane M-protein consist
of an N-terminal ectodomain and a C-terminal endodomain. The endodomain inter-
acts with multiple regions of both the N-protein and S-protein for oligomerization,
RNA encapsulation and mature virus particle formation but also with the E-protein
through two transmembrane and the cytoplasmic domains (Hsieh, Li, Chen, & Lo,
2008). The interactions of the M-protein with the S-protein, E-protein and N-protein
of SARS-CoV-2 have been recently modeled using protein-protein docking and mo-
lecular dynamic simulation (Kumar, Kumar, Garg, & Giri, 2021). The M protein acts
as receptor, while the E-protein, N-protein and S-protein act as protein ligands. For
example, transient helices in the domains and linkers of the N-protein establish in-
teractions with a number of proteins, including the M-protein (Lu et al., 2021) and
NSP3 of the viral-replicase transcription complex (Hurst, Koetzner, & Masters,
2013). Clearly, the haplotype-mediated network of proteins shown in Fig. 6 makes
these interactions evident in VOC evolution.
FIG. 7
Mutations of the S-protein characteristic of VOC omicron cluster in groups according to
haplotype and are enriched in immune evasion functions associated with the RBD region. The
diagram maps mutations onto the different regions of the S-protein molecule from N-terminus
to C-terminus in the amino acid sequence, with markers specific to VOCs alpha and delta
indicated in the top and those specific for VOC omicron in the bottom. Clusters 1, 2 and 3
represent mutations arising from nucleotide substitutions at codon sites that are either
negatively selected or are evolving under no detectable selection in non-omicron sequences.
SP, signal peptide; NTD, N-terminal domain; RBD, receptor-binding domain; RBM, receptor-
binding motif; CS, cleavage site; FP, fusion peptide; IFP, internal fusion peptide; HR1, heptad
repeat 1; HR2, heptad repeat 2; TM, transmembrane domain.
(i) VOC alpha appeared during the second phase of environmental sensing but
carried with it markers typical of protein flexibility/rigidity. It did so after
recruiting the D614G mutation of haplotype 5, which was already highly
prevalent during the first wave of the pandemic. In silico modeling and cryo-
EM-based conformational dynamic studies showed that D614G disturbed
neighboring hydrogen bonding interactions between the S1 and S2 subunits of
pairs of protomers of the spike and contacts with the fusion peptide (FP) region
(Korber et al., 2020; Xu et al., 2021). Population genetic analysis indicated that
D614G provided a selective advantage associated with higher viral loads and
younger age of patients (Voltz et al., 2021). Note that recruitment of Haplotype
5 was soon followed by haplotype 2, which impacted the intrinsically
disordered linker of the N-protein (Tomaszewski et al., 2020). Besides
recruiting these flexibility-associated haplotypes, VOC alpha developed two
main haplotypes affecting spike functionality. The core haplotype 1 involved
A570D, T716I, S982A and D1118H, all of them altering regions of increased
protein disorder (see Fig. 6B in Caetano-Anolles et al., 2022) in domains of the
C-terminal S2 subunit. In turn, haplotype 3 involved three deletions, H69del,
V70del, and Y144del, which were all located in the NTD region holding a
galectin-like structure associated with viral seasonality. The two free-standing
S-protein markers, N501Y and P681H were the first variants to impact the RBD
region responsible for binding ACE2 and other crucial ligands. The N501Y
marker makes up one of 6 key contact residues of RBD shown to increase both
ACE2 receptor affinity and infectivity and virulence (Starr et al., 2020).
254 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
The P681H marker alters one of 4 residues comprising the insertion that creates
the S1/S2 furin cleavage site between the S1 and S2 subunits (see Harvey et al.,
2021). All of these haplotypes and markers were later recruited by VOC
omicron, demonstrating their functional centrality.
(ii) VOC delta appeared during the beginning of the immune escape phase. Its
haplotypes did not retain mutations in the S-protein, except for D614G of
haplotype 5. Instead, it added a large number of mutations in other proteins. Only
two of its 6 haplotypes contained S-protein markers; haplotype 7 contained T19R
and D950N and haplotype 8 contained E156G, F157del and R158del. All of these
markers are located in the environmental sensing NTD region. Free-standing
markers were also NTD-enriched (T96I, G142D, A222V) but contained markers
in the immunogenic RBD region (T478K, L452R) and the furin cleavage site
(P681R). Some of these are the only markers shared with VOC omicron (Fig. 5)
(iii) VOC omicron is now displacing other VOCs at a worldwide level after
recruiting markers from VOC alpha rather than VOC delta and developing a
large marker constellation by haplotype coalescence. However, the massive
acquisition of mutations in the RBD region (with 3 haplotypes) is balanced by
mutations in the NTD and the S2 subunit regions, with 3 and 2 haplotypes,
respectively (Fig. 7). This indicates a significant commitment to immune
escape as we enter the endemic phase of COVID-19 prevalence.
markers but at low prevalence levels (10% levels) in regions 1–4. We postulate all
these markers are sensor proteins recruited by VOCs delta and omicron. Their accu-
mulation support the genetic differences we detected between New South Wales and
Victoria in Fig. 2.
The temporal accumulation of the N501Y and P681H markers of the S-protein
that are shared by VOCs alpha and omicron was also interesting. Prevalence peaked
during the fourth quarter of 2020 reaching 60–100% levels in South Australia and
ACT, disappearing in the third quarter of 2021 and quickly increasing with the rise
of VOC omicron. Again, accumulation in regions 1–4 was in general lower, with the
exception of Queensland. Out of all 6 markers, T478K was the first to reach and
maintain 100% prevalence levels, doing so in the second quarter of 2021 at a time
VOC delta was just starting to accumulate in the planet. All of these rather noisy
patterns of accumulation suggest VOCs engaged in latitude-dependent recruitment
of markers that were already significantly present in the early viral population.
7 Discussion
A recent opinion article claims there is no “transparent” path of transmission linking
VOC omicron to its predecessors and no explanation for the unusual array of muta-
tions that appeared to have evolved outside scrutiny of researchers (Mallapaty,
2022). Indeed, VOC omicron was able to quickly develop a large constellation of
mutations affecting not only the S-protein but also many other proteins of functional
significance (Viana et al., 2022). Similarly, its appearance out of thin air was unan-
ticipated and puzzling. Our results, however, do show that many of VOC omicron
mutations were already present during the first year of the pandemic. They were
recruited piecemeal a year later to be part of a complex mutant constellation. In fact,
16 S-protein variants of VOC omicron, 7 of VOC delta, and all variants of VOC alpha
were already present before November 2020 forming for example combinations of
10 variants in a genome sequence. Some of these mutant combinations were present
in large numbers in the viral population that was sampled for sequencing. These re-
sults strongly support the existence of massive viral recruitments occurring already
during the first waves of the pandemic. The fact that we found that the molecular
profile of VOC omicron did not appear monolithically in different regions of
Australia also challenges two other theoretical explanations for the origin of VOC
omicron (Mallapaty, 2022), namely that all mutations evolved in one patient as part
of a long-term infection or that their emergence occurred unseen in other animal
hosts (e.g., rodents). These scenarios, which are more compatible with evolutionary
founder effects and super-spreader events are therefore unlikely.
VOC omicron was first detected by a genomic surveillance team in November
2021 after a resurgence of infections in the Gauteng Province of South Africa. By
mid-December, this new VOC was present in 87 countries in patterns that
suggested rapid transmission in regions with high levels of population immunity.
7 Discussion 259
by new mutations, especially in the S-protein. For example, after the initial emergence
of the virus, the second wave was predominantly led by two sublineages, B.1 and
B.1.1, and interestingly, its prevalence pattern was geographically diverse with higher
rates of prevalence observed in the North of England compared to the South. Similarly,
the third wave was driven by VOC alpha. With every new epidemic wave, a new
lineage and mutations came to the forefront, which helped the virus escape previous
immunity. A number of refractory variants containing the E484K mutation of the
S-protein (including VOCs beta, gamma, eta, iota and zeta) followed the demise of
VOC alpha and preceded the rise of VOCs kappa and delta. These lineages introduced
a number of new mutations, which were then replaced by the VOC delta constellation.
Both studies suggest that the SARS-CoV-2 genome is evolving dynamically,
with bursts of mutation accumulation followed by sweeps. Patterns of sub-epidemic
waves and the rise of VOCs suggest major global shifts in the selective landscape and
possible convergence between lineages (Martin et al., 2022). In addition, the viral
genome appears to undergo evolutionary change in response to its host, even under
implementation of selective pressures such as vaccination. Mutational changes are
particularly exacerbated when super spreader events drive infrequent mutations to
prominence (Choi et al., 2020).
Our analysis of SARS-CoV-2 genomes in Australia is in line with findings for the
United States and England. Mutations accumulated in bursts while haplotypes were
generated in waves. Two haplotypes were the first to accumulate during the initial
wave of the pandemic early in 2020, haplotype 5 (involving the S-protein and
NSP12) and haplotype 2 (involving the N-protein). The rise of VOC alpha early
in 2021 took advantage of these two haplotypes but also generated an additional
three, haplotype 1 (involving the S-protein, N-protein, ORF8 and NSP3), haplotype
3 (involving the S-protein) and haplotype 4 (involving NSP6). The rise of VOC delta
mid-2021 displaced most markers of VOC alpha but retained haplotype 5. Instead it
generated 6 additional haplotypes of its own, which were highly enriched in markers
of the S-protein. Remarkably, the arrival of VOC omicron at the end of 2021 dis-
placed most markers from VOC delta but retained haplotype 5, recruited the van-
ished haplotypes 2, 3 and 4 and two free-standing S-protein markers of VOC
alpha, and 3 S-protein markers and one NSP4 marker from VOC delta. The three suc-
cessive waves involved 24, 35 and 59 markers, showcasing the gradual structuring of
the mutational landscape of the viral population.
regions of Australia. This suggests there is a global emergence process that depends
on the regional environment and is developing independently of selective sweeps.
To illustrate, Fig. 3A shows the noisy appearance of the two amino acid variants
of haplotype 5, the oldest and most stable haplotype. Markers did not accumulate
uniformly in the different regions as their prevalence was approaching 100%. They
did so idiosyncratically as shown by accumulation curves for the two N-protein
markers of haplotype 2 (Fig. 3B) Their fate was more haphazard and never reached
100% prevalence levels in regions closer to the Equator. In contrast, other haplotypes
did not decouple but showed distinct accumulation patterns in the different regions
(e.g., haplotype 1 of VOC alpha; Fig. 4A).
The atomization of mutant constellations into haplotypes was unanticipated and
shows VOCs are highly dynamic entities that are capable of adding and eliminating
markers from their individual makeup. However, there is some structure to the as-
sembly of haplotypes. When there is no recombination or rearrangement, haplotype
structure reflects the age distribution of mutations in an evolving phylogenetic tree of
viral variants. In the presence of horizontal processes of genetic exchange, every se-
quence site that is subject to mutation can follow a tree but site-specific trees must be
reconciled. In other words, horizontal exchange shuffles genetic variation. This com-
plicates standard views of population genetics, such as forward-in-time evolution of
allele frequencies and backward-in-time genealogical models. One example is the
duality between the Wright-Fisher diffusion for genetic drift and its genealogical
counterpart, the coalescent, which has been only recently modeled to incorporate
the effects of recombination (Griffiths, Jenkins, & Lessard, 2016). Here, an ancestral
recombination graph must be reconciled with genetic drift by diffusion in a land-
scape of mutations in an attempt to explain how ancestral genetic material is dis-
persed across ancestors of a contemporary population. In the case of VOCs of a
viral quasispecies, we have little understanding of these processes. Instead, we
can define haplotypes experimentally by detecting congruent patterns of accumula-
tion of their markers. Patterns that are similar suggest a “coalescence” of haplotypes
or free-standing markers into a same regional behavior. We exploit this coalescence
in an “haplotype graph” that describes how haplotypes share markers and similarities
in patterns of accumulation across regions of Australia (Fig. 5). Three clear sub-
graphs describing the three major VOCs that appeared in Australia are unified by
the universally present haplotype 5 that contains the D614G marker of the spike.
Other haplotypes and free-standing markers also unify VOC makeup. Haplotype
coalescence is described by core haplotypes acting as hubs.
depend strongly on environmental factors, host susceptibility, and the type and level
of immune response that the host mounts against viral infection. Direct and indirect
contact play a significant role in transmission of respiratory viruses. Airborne trans-
mission for example is greatly affected by susceptibility, weather, temperature,
topography, and air quality, all of which contribute to seasonal behavior (Pica &
Bouvier, 2012). In addition, the host’s environment (e.g., time spent indoors), host
defense (e.g., impaired mucociliary clearance through cold and dry air inhalation),
and changes in viral infectability and stability along with climatic conditions are
known to impact the seasonal behavior of respiratory viruses (Tamerius et al.,
2011). Viral transmissibility can also be very high during initial stages of an epi-
demic due to lower immunity, and environmental factors cannot stop transmission
at times (Grenfell & Bjørnstad, 2005). In the case of the influenza virus, temperature
and relative humidity have the greatest impact on seasonality (Lowen, Mubareka,
Steel, & Palese, 2007; Pica & Bouvier, 2012).
The effect of seasonal variations on the transmissibility of SARS-CoV-2 has
become a topic of great interest (Kissler, Tedijanto, Goldstein, Grad, & Lipsitch,
2020). Seasonality warrants attention because it could assist in formulating informed
actionable responses to the COVID-19 pandemic. Emerging evidence suggests that
an association exists between seasonal variations and the survival and transmissibil-
ity of SARS-CoV-2, with higher latitude, colder temperatures, and lower humidity
levels being associated with higher incidence of COVID-19 (Burra et al., 2021; Liu
et al., 2021; Sajadi et al., 2020). Considering the structure of SARS-CoV-2, it is log-
ical that the virus could be sensitive to environmental conditions. The viral capsid
that encases the proteins and genetic material is surrounded by a lipid bilayer
likely to be affected by environmental conditions like temperature and humidity
(Moriyama, Hugentobler, & Iwasaki, 2020). In fact, our research suggests that the
seasonality of COVID-19 can be explained by the presence of a galectin-like struc-
ture in the NTP region of the S1 subunit of the spike protein (Caetano-Anolles et al.,
2022). The NTD, together with the RBD region, facilitates the viral attachment to
cells by recognizing and binding to sugars and other receptors of the host cell
(Pourrajab, 2021). Galectins are a family of evolutionarily conserved effector pro-
teins that regulate a wide range of biological processes including pre-mRNA splicing
and various kinds of cellular interactions, as well as pathogen recognition and in-
flammatory responses (Dings, Miller, Griffin, & Mayo, 2018). These diverse roles
appear to mostly involve binding to the carbohydrate moieties of glycoconjugates
present on the surface of cells (Caetano-Anolles et al., 2022). The presence of cel-
lular structures similar to galectins in the viral spike protein suggests they play a role
in helping SARS-CoV-2 attach to host cells by using the same carbohydrate recep-
tors used by galectins when evading host immune responses (possibly by masking
itself from host galectins; Pourrajab, 2021). Such hypotheses about the role of these
galectin like structures in SARS-CoV-2 have been corroborated by findings on the
administration of SARS-Cov-2 galectin like inhibitors, which reportedly decrease
SARS-CoV-2 loads in patients (Sethi, Sanam, Munagalasetty, Jayanthi, & Alvala,
2020). Remarkably, a relationship between temperature shifts and the activity of
264 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
7.4 Conclusions
Recent analyses of accumulating genome sequence data suggest that the SARS-CoV-2
virus is evolving in bursts while developing with each new VOC an increasing con-
stellation of mutations. This indicates that the virus is furthering persistence by adapt-
ing to the external environment and to the physiology and behavior of its human hosts
(Caetano-Anolles et al., 2022). Although most of the mutations are evolutionarily neu-
tral, VOC markers are under strong positive selection and their haplotype structure
suggest this is occurring by fostering beneficial intramolecular and/or intermolecular
interactions. A rate of 25 amino acid substitutions per year has been reported (esti-
mated with a molecular clock) and the latest VOC omicron shows at least 37 mutations
in the spike protein alone (Cameroni et al., 2021; V€ohringer et al., 2021). The increase
References 265
References
Artika, I. M., Dewantari, A. K., & Wiyatno, A. (2020). Molecular biology of coronaviruses:
Current knowledge. Heliyon, 6(8), e04743.
Bai, Z., Cao, Y., Liu, W., & Li, J. (2021). The SARS-CoV-2 nucleocapsidprotein and its role in
viral structure, biological functions, and a potential target for drug or vaccine mitigation.
Viruses, 13, 1115.
Becerra-Flores, M., & Cardozo, T. (2020). SARS-CoV-2 viral spike G614 mutation exhibits
higher case fatality rate. International Journal of Clinical Practice, 74, e13525.
Burra, P., Soto-Dı́az, K., Chalen, I., Gonzalez-Ricon, R. J., Istanto, D., & Caetano-Anolles, G.
(2021). Temperature and latitude correlate with SARS-CoV-2 epidemiological variables but
not with genomic change worldwide. Evolutionary Bioinformatics, 19. 1176934321989695.
Caetano-Anolles, K., Hernandez, N., Mughal, F., Tomaszewski, T., & Caetano-Anolles, G.
(2022). The seasonal behaviour of COVID-19 and its galectin-like culprit of the viral
spike. Methods in Microbiology, 50, 27–81. https://doi.org/10.1016/bs.mim.2021.10.002.
Cameroni, E., Bowen, J. E., Rosen, L. E., Saliba, C., Zepeda, S. K., et al. (2021). Broadly neu-
tralizing antibodies overcome SARS-CoV-2 omicron antigenic shift. Nature, 602,
664–670.
Campbell, F., Archer, B., Laurenson-Schafer, H., Jinnai, Y., Konings, F., et al. (2021). In-
creased transmissibility and global spread of SARS-CoV-2 variants of concern as at
2021. Euro Surveillance, 26(24), 2100509.
Caswell, T. A., Droettboom, M., Lee, A., et al. (2021). Matplotlib/Matplotlib, v3.5.1. Zenodo.
https://doi.org/10.5281/zenodo.5773480.
Choi, B., Choudhary, M. C., Regan, J., Sparks, J. A., Padera, R. F., et al. (2020). Persistence
and evolution of SARS-CoV-2 in an immunocompromised host. New England Journal of
Medicine, 383(23), 2291–2293.
Cottam, E. M., Maier, H. J., Manifava, M., Vaux, L. C., Chandra-Schoenfelder, P., et al.
(2011). Coronavirus nsp6 proteins generate autophagosomes from the endoplasmic
reticulum via an omegasome intermediate. Autophagy, 7, 1335–1347.
266 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
Davies, N. G., Abbott, S., Barnard, R. C., Jarvis, C. I., Kucharski, A. J., et al. (2021). Estimated
transmissibility and impact of SARS-CoV-2 lineage B.1.1.7 in England. Science,
372(6538). eabg3055.
Deyle, E. R., Maher, M. C., Hernandez, R. D., Basu, S., & Sugihara, G. (2016). Global
environmental drivers of influenza. Proceedings of the National Academy of Science of
the USA, 113, 13081–13086.
Dings, R. P., Miller, M. C., Griffin, R. J., & Mayo, K. H. (2018). Galectins as molecular targets
for therapeutic intervention. International Journal of Molecular Sciences, 19(3), 905.
Domingo, E., Sheldon, J., & Perales, C. (2012). Viral quasispecies evolution. Microbiology
and Molecular Biology Reviews, 76, 159–216.
Eccles, R. (2005). Understanding the symptoms of the common cold and influenza. The Lancet
Infectious Diseases, 5(11), 718–725.
Elbe, S., & Buckland-Merrett, G. (2017). Data, disease and diplomacy: GISAID’s innovative
contribution to global health. Global Challenges, 1, 33–46.
Grenfell, B., & Bjørnstad, O. (2005). Epidemic cycling and immunity. Nature, 433(7024),
366–367.
Griffiths, R. C., Jenkins, P. A., & Lessard, S. (2016). A coalescent dual processes for a Wright-
fisher diffusion with recombination and its application to haplotype partitioning. Theoretical
Population Biology, 112, 126–138.
Harvey, W. T., Carabelli, A. M., Jackson, B., Gupta, R. K., Thomson, E. C., et al. (2021).
SARS-CoV-2 variants, spike mutations and immune escape. Nature Reviews Microbiol-
ogy, 19, 409–424.
Hodcroft, E. B., Hadfield, J., Neher, R. A., & Bedford, T. (2020). Year-letter genetic clade
naming for SARS-CoV-2 on Nextstrain.org. Nextstrain. https://nextstrain.org/blog/
2020-06-02-SARSCoV2-clade-naming.
Hope-Simpson, R. E. (1981). The role of season in the epidemiology of influenza. Journal of
Hygiene (London), 86(1), 35–47.
Hsieh, Y.-C., Li, H.-C., Chen, S.-C., & Lo, S.-Y. (2008). Interactions between M protein and
other structural proteins of severe, acute respiratory syndrome-associated coronavirus.
Journal of Biomedical Science, 15(6), 707–717.
Hunter, J. D. (2007). Matplotlib: A 2D graphics environment. Computing in Science &
Engineering, 9(3), 90–95.
Hurst, K. R., Koetzner, C. A., & Masters, P. S. (2013). Characterization of a critical interaction
between the coronavirus nucleocapsid protein and nonstructural protein 3 of the viral
replicase-transcriptase complex. Journal of Virology, 87, 9159–9172.
Khare, S., Gurry, C., Freitas, L., Schultz, M. B., Bach, G., et al. (2021). GISAID’s role in
pandemic response. CCDC Weekly, 3(49), 1049–1051.
Kissler, S. M., Tedijanto, C., Goldstein, E., Grad, Y. H., & Lipsitch, M. (2020). Projecting the
transmission dynamics of SARS-CoV-2 through the postpandemic period. Science,
368(6493), 860–868.
Korber, B., Fischer, W. M., Gnanakaran, S., Yoon, H., Theiler, J., et al. (2020). Tracking
changes in SARS-CoV-2 spike: Evidence that D614G increases infectivity of the
COVID-19 virus. Cell, 182, 812–827.
Kumar, P., Kumar, A., Garg, N., & Giri, R. (2021). An insight into SARS-CoV-2 membrane
protein interaction with spike, envelope, and nucleocapsid proteins. Journal of Biomolec-
ular Structure and Dynamics. https://doi.org/10.1080/07391102.2021.2016490.
Lauring, A. S., & Hodcroft, E. B. (2021). Genetic variants of SARS-CoV-2—What do they
mean? JAMA, 325(6), 529–531. https://doi.org/10.1001/jama.2020.27124.
References 267
Lei, J., Kusov, Y., & Hilgenfeld, R. (2018). NSP3 of coronaviruses: Structures and functions of
a large multi-domain protein. Antiviral Research, 149, 58–74.
Liu, X., Huang, J., Li, C., Zhao, Y., Wang, D., Huang, Z., et al. (2021). The role of seasonality
in the spread of COVID-19 pandemic. Environmental Research, 195, 110874.
Lowen, A. C., Mubareka, S., Steel, J., & Palese, P. (2007). Influenza virus transmission is
dependent on relative humidity and temperature. PLoS Pathogens, 3(10), e151.
Lu, S., Ye, Q., Singh, D., Cao, Y., Diedrich, J. K., et al. (2021). The SARS-CoV-2 Nucleocapsid
phosphoprotein forms mutually exclusive condensates with RNA and the membrane-
associated M protein. Nature Communications, 12, 502.
Lutomski, C. A., El-Baba, T. J., Bolla, J. R., & Robinson, C. V. (2021). Multiple roles of
SARS-CoV-2 N protein facilitated by proteoform-specific interactions with RNA, host
proteins, and convalescent antibodies. JACS Au, 1, 1147–1157.
Mallapaty, S. (2022). The hunt for the origins of omicron. Nature, 602, 26–27.
Martin, D. P., Lytras, S., Lucaci, A. G., Maier, W., Gr€ uning, B., et al. (2022). Selection
analysis identifies unsusual clustered mutational changes in omicron lineage BA.1 that
likely impact spike function. bioRxiv. https://www.biorxiv.org/content/10.1101/2022.
01.14.476382v1.
Martinez, M. E. (2018). The calendar of epidemics: Seasonal cycles of infectious diseases.
PLoS Pathogens, 14(11), e1007327.
McKinney, W. (2010). Data structures for statistical computing in Python. In S. van der
Walt, & J. Millman (Eds.), Proceedings of the 9th Python in science conference (pp.
56–61).
Moriyama, M., Hugentobler, W. J., & Iwasaki, A. (2020). Seasonality of respiratory viral in-
fections. Annual Review of Virology, 7, 83–101.
Nickbakhsh, S., Ho, A., Marques, D. F., McMenamin, J., Gunson, R. N., & Murcia, P. R.
(2020). Epidemiology of seasonal coronaviruses: Establishing the context for the
emergence of coronavirus disease 2019. The Journal of Infectious Diseases, 222(1),
17–25.
Ogando, N. S., Zevenhoven-Dobbe, J. C., Meer, Y. V. D., Bredenbeek, P. J., Posthuma, C. C.,
Snijder, E. J., et al. (2020). The enzymatic activity of the nsp14 exoribonuclease is critical
for replication of MERS-CoV and SARS-CoV-2. Journal of Virology, 94(23).
e01246–01220.
Pandas Development Team. (2022). Pandas-dev/pandas: Pandas 1.1.5. Zenodo. https://doi.
org/10.5281/zenodo.5893288.
Pica, N., & Bouvier, N. M. (2012). Environmental factors affecting the transmission of
respiratory viruses. Current Opinion in Virology, 2(1), 90–95.
Pourrajab, F. (2021). Targeting the glycans: A paradigm for host-targeted and COVID-19 drug
design. Journal of Cellular and Molecular Medicine, 25(13), 5842–5856.
Rambault, A., Holmes, E. C., O’Toole, A., Hill, V., McCrone, J. T., Ruis, C., et al. (2020).
A dynamic nomenclature proposal for SARS-CoV-2 lineages to assist genomic epidemi-
ology. Nature Microbiology, 5, 1403–1407.
Rodrigues, T. S., de Sá, K. S. G., Ishimoto, A. Y., Becerra, A., Oliveira, S., et al. (2021).
Inflammasomes are activated in response to SARS-CoV-2 infection and are associated
with COVID-19 severity in patients. Journal of Experimental Medicine, 218, e20201707.
Sajadi, M. M., Habibzadeh, P., Vintzileos, A., Shokouhi, S., Miralles-Wilhelm, F., &
Amoroso, A. (2020). Temperature, humidity, and latitude analysis to estimate potential
spread and seasonality of coronavirus disease 2019 (COVID-19). JAMA Network Open,
3, e2011834.
268 CHAPTER 8 The emergence of SARS-CoV-2 variants of concern
Sethi, A., Sanam, S., Munagalasetty, S., Jayanthi, S., & Alvala, M. (2020). Understanding the
role of galectin inhibitors as potential candidates for SARS-CoV-2 spike protein: In silico
studies. RSC Advances, 10(50), 29873–29884.
Showers, W. M., Leach, S. M., Kechris, K., & Strong, M. (2022). Analysis of SARS-CoV-2
mutations over time reveals increasing prevalence of variants in the spike protein and
RNA-dependent RNA polymerase. Infection, Genetics and Evolution, 97, 105153.
Shu, Y., & McCauley, J. (2017). GISAID: Global initiative on sharing all influenza data –
From vision to reality. Eurosurveillance, 22(13), 30494.
Starr, T. N., Greaney, A. J., Hilton, S. K., Ellis, D., Crawford, K. H. D., Dingens, A. S., et al.
(2020). Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals
constraints on folding and ACE2 binding. Cell, 5, 1295–1310.
Sun, X., Huang, Z., Xu, W., & Hu, W., et al. (2021). SARS-CoV-2 non-strucural protein
6 triggers LRRP3-dependent pyroptosis by targeting TP6AP1. Cell Death & Differentia-
tion. https://doi.org/10.1038/s41418-021-00916-7.
Tamerius, J., Nelson, M. I., Zhou, S. Z., Viboud, C., Miller, M. A., & Alonso, W. J. (2011).
Global influenza seasonality: Reconciling patterns across temperate and tropical regions.
Environmental Health Perspectives, 119(4), 439–445.
Tasakis, R. N., Samaras, G., Jamison, A., Lee, M., Paulus, A., et al. (2021). SARS-CoV-2 var-
iant evolution in the United States: High accumulation of viral mutations over time likely
through serial founder events and mutational bursts. bioRxiv. https://doi.org/10.1371/jour-
nal.pone.0255169.
Tomaszewski, T., DeVriers, R. S., Dong, M., Bhatia, G., Norsworthy, M. D., Zheng, X., et al.
(2020). New pathways of mutational change in SARS-CoV-2 proteomes involve regions
of intrinsic disorder important for virus replication and release. Evolutionary Bioinformat-
ics, 16. 1176934320965149.
Viana, R., Moyo, S., Amoako, D. G., Tegally, H., Scheepers, C., et al. (2022). Rapid epidemic
expansion of the SARS-CoV-2 omicron variant in southern Africa. Nature. https://doi.org/
10.1038/s41586-022-04411-y.
V€ohringer, H. S., Sanderson, T., Sinnott, M., De Maio, N., Nguyen, T., et al. (2021). Genomic
reconstruction of the SARS-CoV-2 epidemic in England. Nature, 600(7889), 506–511.
Voltz, E., Hill, V., McCrone, J. T., Proce, A., Jorgensen, D., et al. (2021). Evaluating the
effects of SARS-CoV-2 spike mutation D614G on transmissibility and pathogencity. Cell,
184(1), 64–75.
Williams, T. C., & Burgers, W. A. (2021). SARS-CoV-2 evolution and vaccines: Cause for
concern? The Lancet Respiratory Medicine, 9(4), 333–335.
Wu, Y., Zhou, Z., Wang, J., Luo, J., Wang, L., & Zhang, Y. (2019). Temperature regulates the
recognition activities of a galectin to pathogen and symbiont in the scleractinian coral
Pocillopora damicornis. Developmental & Comparative Immunology, 96, 103–110.
Xu, C., Wang, Y., Liu, C., Zhang, C., Han, W., Hong, X., et al. (2021). Conformational
dynamics of SARS-CoV-2 trimeric spike glycoprotein in complex with receptor ACE2
revealed by cryo-EM. Science Advances, 7(1). eabe5575.
Zhang, Y., Sun, H., Pei, R., Mao, B., Zhao, Z., & Li, H., et al. (2021). The SARS-CoV-2 pro-
tein ORF3a inhibits fusion of autophagosomes with lysosomes. Cell Discovery, 7, 31.
CHAPTER
COVID-19 vaccines
for high risk and
immunocompromised
patients
9
Charles S. Paviaa,b,* and Maria M. Plummerc
a
Department of Biomedical Sciences, NYIT College of Osteopathic Medicine,
New York Institute of Technology, Old Westbury, NY, United States
b
Division of Infectious Diseases, New York Medical College, Valhalla, NY, United States
c
Department of Clinical Specialties, Division of Pathology, NYIT College of Osteopathic Medicine,
New York Institute of Technology, Old Westbury, NY, United States
*Corresponding author: e-mail address: [email protected]
1 Introduction
The COVID-19 pandemic has had a major negative impact on public health and its
worldwide infrastructure and resources. Perhaps the most recognized devastating
outcomes of this crisis are the high rates of morbidity and mortality, and the toll
it has taken on people who have underlying co-morbidities or risk factors (Garg
et al., 2020). Among these, include groups within the general population who are
diabetic (Garg et al., 2020; Gao et al., 2020), obese (de Frel et al., 2020), elderly
(Centers for Disease Control and Prevention, 2021), and possibly immunocom-
promised (Kates et al., 2020; Office of the AIDS Research Advisory Council,
2021), especially people who are infected with the human immunodeficiency virus
(HIV) and who subsequently progress to developing acquired immunodeficiency
disease (AIDS) (Office of the AIDS Research Advisory Council, 2021). Many of
these well-characterized victims have a more severe course of COVID-19, leading
to increased numbers of hospitalized patients and deaths relative to the percentages
occurring in people having little or no pre-conditions (Garg et al., 2020), although,
as will be discussed later on, the pattern in the HIV/AIDS group has been reported
to be less clear and inconsistent (Office of the AIDS Research Advisory Council,
2021). Also included in this latter group having immune defects are transplant
recipients, cancer patients and autoimmune cases where an immunocompromised
or hyperimmune condition leads to intense treatment with certain medications that
adversely affect various components of the immune system. The key reasons why
Methods in Microbiology, Volume 50, ISSN 0580-9517, https://doi.org/10.1016/bs.mim.2021.11.001
Copyright © 2022 Elsevier Ltd. All rights reserved.
269
270 CHAPTER 9 COVID-19 vaccines for high risk and immunocompromised
patients
in this venture, along with the huge response of the general population willing to
participate in clinical trials that were needed to verify the worthiness of these vac-
cines. Even many of the recognized medical/scientific “experts” had predicted that
vaccines would likely not be available for routine use for about 1.5–2 years after the
initiation of experiments in trying to develop them. Certainly, in the back of many
minds was the gloomy feeling that it may even take much longer given the relative
lack of success, after much effort and a large amount of funds had already been
invested, over a span of many years, in trying to develop highly effective vaccines
for malaria, HIV infection and EBOLA virus disease (EVD)—diseases having high
rates of morbidity and mortality (similar outcomes to COVID-19), but which have
been known and well characterized for well over a century (for malaria) and for
nearly a half century (for HIV infection and EVD). On the other hand, the annual
development of vaccines to combat influenza (aka “the flu”) caused by the genetic
variants of the influenza virus did offer encouragement that an effective COVID-19
vaccine could be produced within a relatively short period of time.
In the United States, as of November 2021, two types of anti-COVID-19 vaccines
are in use and available to everyone from ages 5 and above. For several months prior
to this, only people >11 years of age were eligible to receive any one of these vac-
cines based on authorizations made by the U. S. Food and Drug Administration
(FDA) followed by recommendations from the Centers for Disease Control and Pre-
vention (CDC). One type of vaccine consists of purified messenger RNA (mRNA)
given in two intramuscular injections with the 2nd injection given 3–4 weeks after
the first one. In October, 2021, an additional booster injection was recommended by
the FDA and the CDC for those recipients who completed their initial series at least
6 months previously and who are > 65 years of age or have certain other risk factors.
Soon after this announcement was made, the booster recommendation was amended
to include everyone ages 18 and older. The mRNA encodes for the SARS-CoV-2
spike protein. The other vaccine has a viral vector formulation with a live but safe
adenovirus serving as the vector for carrying the genome of the SARS-CoV-2 spike
protein and, until recently, it was thought that a second booster injection would not
be needed for one of the two viral vector versions currently available. However,
serious consideration is now being given to modify this strategy to include additional
boosters for all vaccines with allowances for “mixing and matching” with the mRNA
vaccines based on recommendations by the FDA to be followed by further consid-
eration by the CDC. In both cases, the spike protein had been shown to be highly
immunogenic and the key protective component in pre-clinical studies, and is
associated with a vigorous antibody response. The time frame for these events
was as follows. After an initial Emergency Use Authorization (EUA), that was issued
in December 2020 and then followed by a second one in February 2021 by the FDA,
the two mRNA vaccines produced by Pfizer (Pearl River, NY) and Moderna
(Cambridge, MA) have either received final approval or are pending final
approval for routine use as of late October 2021. This is also true for the viral vector
vaccines produced by Janssen/Johnson & Johnson (Titusville, NJ) and Astra-Zeneca
(Cambridge, UK) which have received an EUA for adults (> 15 year’s old) in the
U.S. It should be noted that in the United Kingdom much earlier approval was given
272 CHAPTER 9 COVID-19 vaccines for high risk and immunocompromised
patients
in December 2020 and January 2021, for some of these vaccines, by the U.K. Health
Department, following recommendations by the Medicare and Health Care Regula-
tory Authority in the Department of Health and Social Care. It is worth noting that,
from the very beginning, the rapid deployment of these vaccines, especially in the
U.S., was partially aided by the involvement of the U.S. federal government under
the program known as “Operation Warp Speed” (United States Federal Government,
2020). Although some people may have viewed this program as having political
overtones, it nonetheless did provide considerable financial and logistical support
to the pharmaceutical industry for accelerating the development and distribution
of multiple vaccine doses throughout much of the U.S. in a nearly unprecedented
and efficient fashion. Also, as an additional benefit of this process, starting in June
2021, surplus vaccine that was being produced in the U.S. (as well as by some of the
other vaccine-producing countries) was then shipped abroad to various countries that
were unable to procure enough vaccine to immunize their citizenry. Still, despite
these humanitarian efforts, in some low-income countries, less than 1% of the pop-
ulation is fully vaccinated, and the success rate is only slightly better in lower-
middle-income countries, compared with more than half in high-income countries.
Thus, such a situation reinforces the ongoing problem of trying to achieve global
vaccine equity. Another related challenge/concern, as pointed out in another chapter
of this volume (Pavia, 2022), is that universal acceptance of the COVID-19 vaccines
is lacking due mostly to hesitancy and the spread of misinformation or a general lack
of a full understanding on the overall benefits of getting vaccinated amid a worldwide
medical disaster. Unfortunately, these anti-vaccine sentiments still linger and will
continue to be deeply engrained in the minds of an ignorant subpopulation and others
unwilling to accept well established facts long after the pandemic has passed.
(i) Are the vaccines safe and what level of protection would these vaccines offer to
this patient population?
(ii) And if so, which vaccine should be administered to them?
(iii) How often should they receive booster injections?
(iv) And at what point, if at all, should they be vaccinated after they have recovered
from a naturally acquired infection with SARS-CoV-2?
3 COVID-19 vaccines for the high-risk patient population 275
The answers to most of these questions are relatively straightforward and not too
complicated. According to the CDC (2020), the U.S. vaccine safety system makes
sure all vaccines are as safe as possible. COVID-19 vaccines have gone through
rigorous safety tests and have met or even exceeded similar standards as for other
vaccines that have been produced for nearly a century and have been in routine
use for over 60 years. People with HIV have been included in clinical trials, though
there is limited safety and efficacy data available as they pertain specifically to this
group. So far, there are no data indicating that the vaccines are not safe and effective
for people with HIV, including adolescents between 12 and 15 years nor has it been
shown that the COVID-19 vaccines interfere with the effectiveness of HIV medica-
tions used in certain treatment regimens, such as HAART. There have been no
unusual links or enhanced negative reactions between HIV or other types of immuno-
suppression with any of the rare serious adverse events that have been reported for
the COVID-19 vaccines within the general population. Much of this information has
been provided by the British HIV Association (2021) and the World Health
Organization (2021), who have also indicated that HIV-infected people generally
will likely produce a weaker response to the COVID-19 vaccines, but they are still
expected to be protected from developing serious illness. This protection, however,
may be to a lesser extent, especially for those individuals with low CD4 + counts
(< 100/mm3). Nonetheless, the U.K. Department of Health recommends that people
with HIV, regardless of their CD4 + count, should receive a COVID-19 vaccine. In
addition, because some people with HIV, especially those with a very low CD4
T-cell count, may be at increased risk for severe illness due to COVID-19, the
CDC recommendation (CDC, 2020) advises that people with HIV may receive
the vaccine as long as they do not have other conditions that would exclude them,
such as a known severe allergic reaction or immediate allergic reaction of any sever-
ity after receiving a previous dose or to a component of the COVID-19 vaccine. The
vaccines authorized for use in the United States and the United Kingdom do not con-
tain live infectious virus so they are expected to be safe in people with low CD4 cell
counts. It is a general rule that people with T-cell defects should not be given live
vaccines, even though they are attenuated, so that they are supposed to only immu-
nize the recipient and not cause disease. However, the attenuated pathogen in such
vaccines could possibly mutate and revert to a more virulent form and cause serious
disease in the immunocompromised host. Examples of this type of vaccine, that are
currently used routinely, include the measles, chickenpox and one of the anti-
shingles vaccines. This same concept applies for the COVID-19 viral vector vaccine
products except they are not “true” live, attenuated vaccines, in the traditional sense.
This is because the available ones now in widespread use, that are produced by
Jannsen/Johnson & Johnson and Astra-Zeneca, consist of a live, but replication-
incompetent, human adenovirus vector, encoding for the recombinant SARS-
CoV-2 spike (S) glycoprotein, stabilized in its pre-fusion form. This differs from
the vaccines produced by Pfizer and Moderna which use only synthetic mRNA as
the immunogen. The viral vector’s purpose is to introduce the DNA, that encodes
for the spike protein, into the human body in a somewhat unique way so that multiple
276 CHAPTER 9 COVID-19 vaccines for high risk and immunocompromised
patients
copies of it can be produced in vivo, over a short period of time, until the crippled
virus immunogen is neutralized by the immune response that is being induced by
the “replicating antigens” (R.J. North, personal communication) of SARS-CoV-2.
This provides a persistent, albeit temporary, stimulus to the vaccinee. Given the
strength and durability of this type of response, this usually means that, theoretically,
and similar to what occurs with a truly live vaccine, a repeat (i.e., a booster) injection
may not be necessary. As noted earlier, this consideration on whether to give boosters
of the viral vector vaccine was being re-evaluated in October 2021 by the U.S. FDA
and CDC, and starting in November 2021, a third (booster) injection is now highly
recommended for most eligible people irrespective of what type of vaccine was
previously given.
Additional features of the viral vector vaccine is that it is considered to be safe
even for immunocompromised patients, including those with HIV/AIDS, given
that the viral vector has been shown to be harmless in one of the most recently pub-
lished studies on this topic (Hammer et al., 2013). Nonetheless, prior to this finding,
there was some initial concern about a potential association observed more than a
decade ago between adenovirus vector-based vaccines and an increased risk of ac-
quiring HIV infection among men who received this type of vaccine (Buchbinder,
McElrath, Dieffenback, & Corey, 2020, 18). This unexpected finding was detected
in two HIV vaccine trials that used adenovirus vector containing products
(Buchbinder et al., 2008; Gray et al., 2011) but these vaccines were constructed
differently and are not related to the structure of the COVID-19 vaccines. The reason
for this previously observed HIV risk remains uncertain, although several follow-up
studies have suggested a possible interference in the HIV-specific vaccine response
or in the CD4 cell susceptibility to HIV infection induced by this kind of vaccine as
an unexpected side effect (Frahm et al., 2012; Perreau, Pantaleo, & Kremer, 2008).
Accordingly, specific studies on this issue with this type of COVID-19 vaccine
should be considered by closely monitoring the response patterns of HIV-infected
people to various immune parameters that would include periodically measuring
CD4 T cell counts, viral loads and anti-spike protein antibody levels subsequent
to being vaccinated. In addition, testing for delayed-type hypersensitivity responses,
as shown recently by Barrios et al. (2021) for recovering COVID-19 patients without
HIV, may also provide valuable insights on the importance of cellular immune re-
sponses mediated by CD8 + T cells that directly kill virally infected cells as being an
additional defense mechanism for prospective vaccine recipients. There are still,
however, other unresolved issues. For example, will the currently available vaccines
be fully protective against variants of SARS-CoV-2, especially the highly invasive/
infectious Delta variant that was first identified in December 2020 (Nature, 2021;
Torjesen, 2021), along with the Omicron variant with its pecular set of mutations
that was recognized and had spread to many countries in the latter part of 2021? What
is the longevity of protection that is provided by any of these vaccines; and will
additional boosters be needed beyond what is currently being done or recommended?
In this regard, it is still unclear or not fully known how much SARS-CoV-2
antigen(s) is needed, or produced in vivo to induce an immune response that leads
to optimal protection against COVID-19. Somewhat encouraging news, along these
References 277
lines, was recently reported (CNN, 2021) indicating that the Janssen/Johnson &
Johnson vaccine provided lasting protection of at least 8 months duration and it
afforded protection against the Delta and other variants. Although there had already
been prior preliminary evidence (reviewed in Chen & Wherry, 2020) supporting an
important role for both CD4 + and CD8 + T cells in the immunologic memory com-
ponent in the host response to COVID-19, it is likely that additional related news on
these topics will be forthcoming in the coming months. In light of these potential
concerns/issues, it should be realized that, in the final analysis, the overall benefits
of receiving any of the authorized COVID-19 vaccines in a pandemic situation
currently outweigh the potential risks, even for people with impaired immune sys-
tems or other co-morbid conditions.
Funding
None to declare.
Author contribution
The authors equally contributed to the conceptualization and writing of the manuscript.
Conflict of interest
The authors declare that they have no conflicts of interest. It should be noted that the citing of
commercially available vaccine products should not be construed as an endorsement. They are
being cited for the sole purpose of providing examples of vaccines that are in either the near-
approval stage, have been approved or been given preliminary authorization for use for im-
munization purposes by authorizing/governmental agencies, after the manufacturers provided
data showing that they have met the minimal standards for successfully completing and
fulfilling the required clinical trial testing parameters for safety and efficacy.
References
Barrios, Y., Franco, A., Sanchez-Machin, I., Poza-Guedes, P., Gonzalez-Perez, R., &
Matheu, V. (2021). A novel application of delayed-type hypersensitivity reaction to mea-
sure cellular immune response in SARS-CoV-2 exposed individuals. Clinical Immunol-
ogy, 226, 108730. https://doi.org/10.1016/j.clim.2021.108730/.
Berbudi, A., Rahmadika, N., Tjahjadi, A. I., & Ruslami, R. (2020). Type 2 diabetes and its
impact on the immune system. Current Diabetes Reviews, 16(5), 442–449. https://doi.
org/10.2174/1573399815666191024085838.
Bertagnolio, S., Thwin, S. S., Silva, R., Ford, N., Baggaley, R., Vitoria, M., et al. (2021).
Clinical characteristics and prognostic factors in people living with HIV hospitalized
with COVID-19: Findings from the WHO Global Clinical Platform. July 2021. https://
theprogramme.ias2021.org/Abstract/Abstract/2498.
Boulle, A., Davies, M. A., Hussey, H., Ismail, M., Morden, E., et al. (2020). Risk factors for
COVID-19 death in a population cohort study from the Western Cape Province, South
Africa. Clinical Infectious Diseases, ciaa1198. https://doi.org/10.1093/cid/ciaa1198.
278 CHAPTER 9 COVID-19 vaccines for high risk and immunocompromised
patients
British HIV Association. 2021 SARS-CoV-2 vaccine advice for adults living with HIV:
British HIV Association (BHIVA) & Terrence Higgins Trust (THT) guidance. Available
online: https://www.bhiva.org/SARS-CoV-2-vaccine-advice-for-adults-living-with-HIV-
update. 11 January 2021.
Brown, L. B., Spinelli, M. A., & Gandhi, M. (2021). The interplay between HIV and
COVID-10: Summary of the data and responses to date. Current Opinion in HIV & AIDS,
16, 63–73. https://doi.org/10.1097/COH.0000000000000659.
Buchbinder, S. P., McElrath, M. J., Dieffenback, C., & Corey, L. (2020). Use of adenovirus
type-5 vectored vaccines: A cautionary tale. Lancet, 396(10260), E68–E69. Available
online: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(20)32156-5/
fulltext#articleInformation.
Buchbinder, S. P., Mehrotra, D. V., Duerr, A., Fitzgerald, D. W., Mogg, R., Li, D., et al.
(2008). Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study):
A double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet, 372,
1881–1893.
Centers for Disease Control and Prevention. 2020. COVID-19 and HIV. Available online:
https://www.cdc.gov/hiv/basics/covid-19.html. 20 October 2020.
Centers for Disease Control and Prevention. 2021. COVID-19 risks and vaccine information
for older adults. https://www.cdc.gov/aging/covid19/covid19-older-adults.html. Updated
1 August 2021.
Chen, Z., & Wherry, E. J. (2020). T cell response in patients with COVID-19. Nature Reviews
Immunology, 20, 529–535. https://doi.org/10.1038/s41577-020-0402-6.
CNN. 2021 J & J Covid-19 vaccine lasts at least 8 months, protects against Delta variant,
studies find. Available online: https://www.cnn.com/2021/07/01/health/johnson-vaccine-
delta-variant/index.html. 1 July 2021.
de Frel D, et al. 2020. The impact of obesity and lifestyle on the immune system and suscep-
tibility to infections such as COVID-19. Frontiers in Nutrition. Nov 19;7:597600,
eCollection 2020. https://doi.org/10.3389/fnut.2020.597600.
Frahm, N., DeCamp, A. C., Friedrich, D. P., Carter, D. K., Defawe, O. D., Kubin, J. G., et al.
(2012). Human adenovirus-specific T cells modulate HIV-specific T cell responses to an
Ad5-vectored HIV-1 vaccine. The Journal of Clinical Investigation, 122, 359–367.
Gao, F., Zheng, K. I., Wang, X. B., Sun, Q. F., Pan, K. H., Wang, T. Y., et al. (2020). Obesity is
a risk factor for greater COVID-19 severity. Diabetes Care, 43, e72–e74. https://doi.org/
10.2337/dc20-0682.
Garg, S., Kim, L., Whitaker, M., O’Halloran, A., Cummings, C., Holstein, R., et al. (2020).
Hospitalization rates and characteristics of patients hospitalized with laboratory-
confirmed coronavirus disease 2019. COVID-NET, 14 states. Morbidity and Mortality
Weekly Report, 69, 458–464. https://doi.org/10.15585/mmwr.mm6915e3.
Gray, G. E., Allen, M., Moodie, Z., Churchyard, G., Bekker, L. G., Nchabeleng, M., et al.
(2011). Safety and efficacy of the HVTN 503/Phambili study of a clade-B-based
HIV-1 vaccine in South Africa: a double-blind, randomized, placebo-controlled test-of-
concept phase 2b study. The Lancet. Infectious Diseases, 11, 507–515.
Hammer, S. M., Sobieszczyk, M. E., Janes, H., Karuna, S. T., Mulligan, M. J., Grove, D., et al.
(2013). Efficacy trial of a DNA/rAd5 HIV-1 preventive vaccine. The New England
Journal of Medicine, 369, 2083–2092.
Johnston, R. (2021). The first 6 months of HIV-SARS-CoV-2 coinfection: Outcomes for
6947 individuals. Current Opinion in HIV & AIDS, 16, 54–62. https://doi.org/10.1097/
COH.0000000000000654.
References 279
Kates, O. S., et al. (2020). Coronavirus disease 2019 in solid organ transplant: a multicenter
cohort study. Clinical Infectious Diseases. https://doi.org/10.1093/cid/ciaa1097.
Kumar, V., Abbas, A., & Aster, J. (2020). Robins and cotran pathophysiology of disease
(10th ed.). London, UK: Elsevier.
McLaughlin, T., Ackerman, S. E., Shen, L., & Engleman, E. (2017). Role of innate and
adaptive immunity in obesity-associated metabolic disease. The Journal of Clinical
Investigation, 127(1), 5–13, 5199693. https://doi.org/10.1172/JCI88876. 28045397.
Murray, P. R., Rosenthal, K. S., & Pfaller, M. A. (2020). Medical microbiology (9th ed.).
London, UK: Elsevier.
Nature. Delta coronavirus variant: Scientists brace for impact. 2021. Available online: https://
www.nature.com/articles/d41586-021-01696-3. 22 June 2021.
Office of the AIDS Research Advisory Council. 2021. Guidance for COVID-19 and People
with HIV j NIH. https://clinicalinfo.hiv.gov/en/guidelines/covid-19-and-persons-hiv-
interim-guidance/interim-guidance-covid-19-and-persons-hiv. Updated 26 February 2021.
Pavia, C. (2022). Hesitancy to get vaccinated against COVID-19 and how it might be over-
come. In C. S. Pavia & V. Gurtler (Eds.), 50. Methods in Microbiology, COVID-19:
Biomedical Perspectives (pp. 223–231). London: UK: Elsevier.
Perreau, M., Pantaleo, G., & Kremer, E. J. (2008). Activation of a dendritic cell-T cell axis
by Ad5 immune complexes creates an improved environment for replication of HIV
in T cells. The Journal of Experimental Medicine, 205, 2717–2725.
Richard, C., Wadowski, M., Goruk, S., et al. (2017). Individuals with obesity and type 2
diabetes have additional immune dysfunction compared with obese individuals who are
metabolically healthy. BMJ Open Diabetes Research & Care, 5, e000379. https://doi.
org/10.1136/bmjdrc-2016-000379.
Saag, M. S. (2021). HIV infection—Screening, diagnosis, and treatment. The New England
Journal of Medicine, 384, 2131–2143.
Torjesen, I. (2021). Covid-19: Delta variant is now UK’s most dominant strain and spreading
through schools. BMJ, 373. https://doi.org/10.1136/bmj.n1445, n1445.
United States Federal Government. 2020 Operation Warp Speed. Available online: https://
www.defense.gov/Explore/Spotlight/Coronavirus/Operation-Warp-Speed/. 15 May 2020.
World Health Organization. 2021 Coronavirus disease (COVID-19): COVID-19 vaccines and
people living with HIV. Available online: https://www.who.int/news-room/q-a-detail/
coronavirus-disease-(covid-19)-covid-19-vaccines-and-people-living-with-hiv. 14 July 2021.
This page intentionally left blank