ArticlePDF AvailableLiterature Review

Abstract and Figures

COVID-19 has engulfed the world and it will accompany us all for some time to come. Here, we review the current state at the milestone of 1 year into the pandemic, as declared by the WHO (World Health Organization). We review several aspects of the on-going pandemic, focusing first on two major topics: viral variants and the human genetic susceptibility to disease severity. We then consider recent and exciting new developments in therapeutics, such as monoclonal antibodies, and in prevention strategies, such as vaccines. We also briefly discuss how advances in basic science and in biotechnology, under the threat of a worldwide emergency, have accelerated to an unprecedented degree of the transition from the laboratory to clinical applications. While every day we acquire more and more tools to deal with the on-going pandemic, we are aware that the path will be arduous and it will require all of us being community-minded. In this respect, we lament past delays in timely full investigations, and we call for bypassing local politics in the interest of humankind on all continents.
SARS-CoV-2 genomic surveillance. a Phylogenetic tree of 19,438 SARS-CoV-2 genomes. Up to December 31, 2020, more than 295,000 complete genomes were submitted to the GISAID database. Each circle in this tree represents a genome that was sequenced over time, since the beginning of the pandemic. As SARS-CoV-2 establishes new infections, its descendants form lineages of genetically related viruses, which can circulate more locally, as shown in lineages represented by threads of circles with similar colors (as shown in red, at the top of panel a), or may have more heterogeneous distributions, as depicted in lineages with multiple colors, highlighting the exchange of viruses between geographic regions (as shown at the bottom of panel a). b State level distribution of SARS-CoV-2 genomes shown in panel a (see acknowledgement Table S1). All regions of the world were impacted by the pandemic, some more than others. As a result, an imbalance in the distribution of genomes worldwide (depicted in panel b as bubbles of distinct sizes) is evident. The differences in genome sampling across continents and countries may not only be the result of epidemic control via distinct public health strategies (as observed in New Zealand and Australia, for example) but may also result from socioeconomic disparities at national and international scales, where some regions (e.g., South and Central America, most regions in Africa), despite being hard hit by the pandemic, are unable to conduct genomic surveillance at a scale comparable to that of rich countries in North America and Europe. Analyses and illustrations were respectively generated using augur and auspice (by nextstrain.org) [11]
… 
This content is subject to copyright. Terms and conditions apply.
R E V I E W Open Access
COVID-19 one year into the pandemic:
from genetics and genomics to therapy,
vaccination, and policy
Giuseppe Novelli
1,2,3*
, Michela Biancolella
4
, Ruty Mehrian-Shai
5
, Vito Luigi Colona
1
, Anderson F. Brito
6
,
Nathan D. Grubaugh
6
, Vasilis Vasiliou
7
, Lucio Luzzatto
8
and Juergen K. V. Reichardt
9
Abstract
COVID-19 has engulfed the world and it will accompany us all for some time to come. Here, we review the current
state at the milestone of 1 year into the pandemic, as declared by the WHO (World Health Organization). We
review several aspects of the on-going pandemic, focusing first on two major topics: viral variants and the human
genetic susceptibility to disease severity. We then consider recent and exciting new developments in therapeutics,
such as monoclonal antibodies, and in prevention strategies, such as vaccines. We also briefly discuss how
advances in basic science and in biotechnology, under the threat of a worldwide emergency, have accelerated to
an unprecedented degree of the transition from the laboratory to clinical applications. While every day we acquire
more and more tools to deal with the on-going pandemic, we are aware that the path will be arduous and it will
require all of us being community-minded. In this respect, we lament past delays in timely full investigations, and
we call for bypassing local politics in the interest of humankind on all continents.
Keywords: Coronavirus, SARS-CoV-2, COVID-19, Pandemic, Variants, Vaccines, Monoclonal antibodies, Politics
Introduction
The coronavirus disease 2019 (COVID-19), caused at in-
dividual level by the severe acute respiratory syndrome
coronavirus 2 (SARS-CoV-2), has raged for a year now,
as declared by the WHO (World Health Organization)
[1]. By March 31, 2021, more than 125 million cases of
SARS-CoV-2 infection have been reported, causing 2,
816,081 deaths in 192 countries (Johns Hopkins Univer-
sity, CSSE). About the Coronavirus Treatment Acceler-
ation Program (CTAP), there are more than 590 drug
development programs in planning stages (i.e., antivirals,
immunomodulators, cell and gene therapies, compound
combinations and other active principles, vaccines
excluded), more than 430 clinical trials reviewed by FDA
(Food and Drug Administration) with a total of 9
COVID-19 treatments currently approved for use under
Emergency Use Authorization (EUA) (https://www.fda.
gov/drugs/coronavirus-covid-19-drugs/coronavirus-
treatment-acceleration-program-ctap, accessed on
March 31, 2021) [2]. Due to socioeconomic inequalities,
such clinical trials are mainly conducted in/by institu-
tions in high income countries. Disparities also impose
large limitations on low/mid income countries, which
are often unable to cope with high demands for molecu-
lar testing, variant surveillance, vaccine distribution, or
to address the needs of their citizens for financial aid, to
survive amid tough epidemic control measures. This
shows that the effects of this pandemic may well be far-
reaching and long-lasting. The central role and responsi-
bility of the UN (United Nations) and specifically of the
WHO are paramount. The very word pandemic means
© The Author(s). 2021 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License,
which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if
changes were made. The images or other third party material in this article are included in the article's Creative Commons
licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons
licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the
data made available in this article, unless otherwise stated in a credit line to the data.
* Correspondence: novelli@med.uniroma2.it
1
Department of Biomedicine and Prevention, Tor VergataUniversity of
Rome, 00133 Rome, Italy
2
IRCCS Neuromed, Pozzilli, IS, Italy
Full list of author information is available at the end of the article
Novelli et al. Human Genomics (2021) 15:27
https://doi.org/10.1186/s40246-021-00326-3
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
that it cannot be confronted by measures that are only
at the level of a region, or of a country, or even of a con-
tinent: global measures are absolutely needed.
On the origin of the COVID-19 pandemic
We note that last year already the WHO set up a panel
to investigate the origin, preparedness, and response to
theCOVID-19pandemic(https://www.who.int/news/
item/09-07-2020-independent-evaluation-of-global-
covid-19-response-announced)[3]. Regrettably, there
has been only limited progress and the origin of the pan-
demic has not been definitively pinned down (https://
www.sciencemag.org/news/2021/03/compromise-who-
report-resolves-little-pandemic-s-origins-details-probe-s-
next-steps)[4]. All of us, whether directly affected pa-
tients or not, should be concerned by such astounding
delays, since it is of immense common interest for all to
be best prepared for the next pandemic that will inevit-
ably befall us eventually. We lament these delays and we
call for expeditious, effective, and scientifically rigorous
action by the WHO and the UN. We are not suggesting
that these delays are politically motivated: but we are
anxious for these organizations to show clear evidence
that they are driven at all times by their feeling of re-
sponsibility toward all humankind. As scientists who are
also citizens from around the world, we wish to be able
to say that they are now acting more nimbly and self-
assuredly. We also wish to highlight the importance of
being mindful that emerging nations that will need
thoughtful assistance in order to face the immediate
health crisis, as well as the economic recovery thereafter:
they need improved and more resilient health systems
on a medium to long-term basis, as well as food security.
In this context, a critical role should be played by other
international organizations such as the EU (European
Union) and notably the FAO (Food and Agriculture
Organization of the UN). However, other regional orga-
nizations, such as ASEAN (Organization of Southeast
Asian States), the AU (African Union), and the OAS
(Organization of American States), should also play a
significant and culture-sensitive role in their respective
geographic areas.
Virus variants
An important reason of concern for all countries is the
emergence of virus variants as a result of mutation(s)
during the current pandemic. RNA viruses, such as
SARS-CoV-2, despite being endowed with proofreading
activity during viral replication [5], have a high mutation
rate, and the absolute number of mutations increases
with every round of infection [6]. The average evolution-
ary rate of SARS-CoV-2 is ~0.8 × 10
4
substitutions per
site per year, which equals about 2 substitutions (muta-
tions) per month. Mutations that are deleterious or
even lethal to the virus will be purged from the popula-
tion, and we do not need to worry about those. Many
mutations are essentially neutral, and are maintained in
the population: they may not readily promote functional
changes, but they may facilitate adaptation upon changes
in the environment explored by viruses [7,8]. However,
a few mutations may be beneficial to the virus [9] be-
cause they lead to (i) increased transmissibility; (ii)
higher infectiousness, (iii) higher virulence entailing
higher rate of severe disease; (iv) immune/vaccine es-
cape; or (v) any combination of the above.
Up to March 31, 2021, nearly 1 million (931,463)
SARS-CoV-2 genomic sequences were submitted to the
GISAID, the main database used by researchers in the
field (https://www.gisaid.org)[10] (Fig. 1, Table S1).
SARS-CoV-2 variants identified are heterogeneously dis-
tributed in geographic areas of the world (see https://
nextstrain.org/ncov/global)[11,12]. Variants of SARS-
CoV-2 may adapt differently to the host under individual
selective pressures [7]; some mutations may increase in
frequency, either through genetic drift or through selec-
tion, and become fixed in different populations [6,7].
This is the case of the mutation that led to the amino
acid change D614G (Asp614 Gly) in the spike glyco-
protein (S), found in the predominant form of SARS-
CoV-2 [13,14]. Patients infected with the D614G variant
often have higher viral loads in the upper respiratory
tract than seen with the ancestral strain, but there seems
to be no difference in disease severity [15]. The D614G
mutation determines an important conformational
change in the spike protein between the S1 and S2 do-
mains, that favors the binding to the angiotensin-
converting enzyme 2 (ACE2) receptor and thus increases
the probability of infection: presumably the key to this
variant having become globally dominant [15]. Recently,
Huang et al. [13] have attributed the selective advantage
of D614G variant to the quantitative differences in
ACE2 expression in different populations. The lower
ACE2 expression observed in the European, North
American, and African populations, compared to Asians,
may have driven positive selection favoring the D614G
mutation.
Many SARS-CoV-2 mutations appeared and were se-
lected for several times, independently, e.g., those that
changes the asparagine residue at spike position 501 (S:
N501Y, S:N501T, S:N501S). This residue is within the
receptor-binding domain (RDB), that is important for
both binding to ACE2 and for antibody recognition. A
variant with S:N501Y, B.1.1.7 (also known as 20B/
501Y.V1), was announced in the South East of England
on December 14, 2020 [14]. Variants from this particular
lineage are associated with multiple amino acid changes
in the spike protein, including a deletion at 69/70 [16],
Y144 deletion, and P681H (adjacent to the furin cleavage
Novelli et al. Human Genomics (2021) 15:27 Page 2 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
site). Rapidly spreading variants were also detected in
South Africa. Viruses belonging to the lineage B.1.351
(also known as 20H/501Y.V2) were detected in Decem-
ber 2020 [17]. These variants are associated with mul-
tiple amino acid changes in spike protein, including S:
N501Y, S:K417N, and S:D80A, but they do not have the
deletion at 69/70 (Table 1). After the detection of these
variants harboring similar genetic changes, genomic sur-
veillance in countries experiencing high COVID-19 inci-
dence started to report more variants with convergent
genetic traits. In late 2020, a new variant was detected in
Manaus, state of Amazonas, northern Brazil [18]. The
new lineage, named P.1 (descendant of B.1.1.28, also
known as 20J/501Y.V3), contains a unique constellation
of lineage-defining mutations, including several amino
acid changes of biological significance known as S:
E484K, S:K417T, and S:N501Y. The P.1 lineage was
identified in 42% (13 of 31) of RT-PCR positive samples
collected between 15 and 23 December 2020, but was
absent in 26 publicly available genome surveillance sam-
ples collected in Manaus between March and November
2020. These results indicate local transmission and pos-
sibly a recent increase in the frequency of a new lineage
from the Amazon region [18]. Finally, two lineages origi-
nated in California, USA, have also emerged and in-
creased in frequency from late 2020 to early 2021,
named B.1.427 and B.1.429, both showing three amino
acid substitutions: S:S13I, S:W152C, and S:L452R [19].
Variants from these two lineages have higher transmissi-
bility (from 18.6 to 24%) when compared with wild type
variants.
Some of the variants of concern have not only been as-
sociated with increased transmission potential but also
with reduced susceptibility to neutralizing antibodies
Fig. 1 SARS-CoV-2 genomic surveillance. aPhylogenetic tree of 19,438 SARS-CoV-2 genomes. Up to December 31, 2020, more than 295,000
complete genomes were submitted to the GISAID database. Each circle in this tree represents a genome that was sequenced over time, since
the beginning of the pandemic. As SARS-CoV-2 establishes new infections, its descendants form lineages of genetically related viruses, which can
circulate more locally, as shown in lineages represented by threads of circles with similar colors (as shown in red, at the top of panel a), or may
have more heterogeneous distributions, as depicted in lineages with multiple colors, highlighting the exchange of viruses between geographic
regions (as shown at the bottom of panel a). bState level distribution of SARS-CoV-2 genomes shown in panel a(see acknowledgement Table
S1). All regions of the world were impacted by the pandemic, some more than others. As a result, an imbalance in the distribution of genomes
worldwide (depicted in panel bas bubbles of distinct sizes) is evident. The differences in genome sampling across continents and countries may
not only be the result of epidemic control via distinct public health strategies (as observed in New Zealand and Australia, for example) but may
also result from socioeconomic disparities at national and international scales, where some regions (e.g., South and Central America, most regions
in Africa), despite being hard hit by the pandemic, are unable to conduct genomic surveillance at a scale comparable to that of rich countries in
North America and Europe. Analyses and illustrations were respectively generated using augur and auspice (by nextstrain.org)[11]
Table 1 Important variants of SARS-CoV-2 that emerged in late 2020
Lineage Other designations Likely origin Key genetic changes
B.1.1.7 20I/501Y.V1 UK 69-70del, 144del, S:N501Y
B.1.351 20H/501Y.V2 South Africa S:E484K, S:N501Y
P.1 20J/501Y.V3 Brazil S:E484K, S:N501Y
Evidence from epidemiological and in vitro assays suggests that variants bearing the key genetic changes listed below are more transmissible
Novelli et al. Human Genomics (2021) 15:27 Page 3 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
from convalescent patients and vaccines (immune scape)
[20]. However, it is important to point out that cellular
response conferred by vaccines is robust, identifies epi-
topes from many proteins beyond the Spike, and major
losses of vaccine efficacy would mainly come as a cumu-
lative effect of several widespread genetic changes that
SARS-CoV-2 undergoes as it continues to spread [21].
Another important consequence of mutations is pheno-
typic changes of virulence. Initial studies have been sug-
gesting that some variants may cause more severe
illness, as already reported for B.1.1.7 [19,22].
The emergence of SARS-CoV-2 variants with concern-
ing phenotypes underscores the importance of genomic
surveillance. The ability to track the spread of variants
differs dramatically across regions, both in international
and national levels [23,24]. To prevent unnoticed viral
spread, and to be able to promptly respond to new vari-
ants as they emerge, genomic surveillance needs to be
incorporated as a routine activity by local departments
of public health, sequencing a minimum percent of re-
ported cases (1-5% of cases or more) per administrative
division (towns, cities, counties, state, etc.).
Human genetic susceptibility
Like in all infectious diseases, while pathogen genetics
plays an important role, host genetics and physiology are
key elements in determining the clinical course of dis-
ease in COVID-19 patients. The main unspecific symp-
toms of the disease are fever, myalgia, fatigue, and dry
cough. As known, SARS-CoV-2 first affects the respira-
tory tract and then activates a systemic inflammatory re-
sponse that can lead to interstitial pneumonia, up to
more critical conditions. The worldwide infection fatality
rate (IFR) is currently estimated around 2-3%, with a
variability depending on different genetic and non-
genetic factors, like sex and age above all [25]. It is a
matter of fact that environmental factors contribute to
the disease severity, but the health status represents a
background that should not be underestimated.
Comorbidities
Virus-host interaction plays a fundamental role in the
diseases outcome. Although most patients have a favor-
able prognosis, some groups are at higher risk, or ex-
tremely vulnerableto severe illness. These include first
and foremost individuals with impaired immune system
function; but also those with cancer, severe lung disease,
such as chronic obstructive pulmonary disease (COPD),
and pregnant women with cardiological disease [26,27].
At high riskare also older patients, very obese individ-
uals with obstructive sleep apnea syndrome (OSAS), and
those with diabetes mellitus, neurological disorders, or
heart, lung, liver, and kidney diseases, who are especially
vulnerable to virus-induced acute respiratory distress
syndrome (ARDS) [28]. This general risk stratification
has exceptions: even young people without comorbidity
may develop severe disease that may even become fatal.
In order to explain this, several hypotheses have been
formulated, including breakdown of immunological tol-
erance, the viral load, an innate immune inefficiency,
and the presence of common or rare risk alleles in genes
encoding proteins important for the biological cycle of
the virus [29,30]. A deeper knowledge of the involve-
ment of the alterations affecting these pathways and the
innate and adaptive immune system may represent a
turning point for understanding the pathophysiological
mechanisms of SARS-CoV-2 and the development of
new therapeutic strategies. Variants in the genes that en-
code these proteins could contribute to different re-
sponses to infections.
Genetic factors
Common and rare variants have been identified in differ-
ent studies using different approaches (Table 2): Genome
wide association studies (GWAS) and deep sequencing in
selected cohorts and/or large biobank resources [35,45
50]. Association studies made it possible to identify in a
number of genes susceptibility alleles for severe disease
phenotypes: however, so far, the risk values are too low
(OR <2) to be regarded as predictive genomic markers
(Table 2). Nonetheless, the additive effects of this low
penetrance alleles might become important in the future
through polygenic scores analysis [51]. On the other
hands, highly penetrance alleles of genes encoding pro-
teins involved in important pathways such as those of in-
nate immunity (e.g., TLR3,IRF7) may be already useful for
risk stratification and potentially useful for prognosis and
treatment [30,31]. A recent (not yet peer-reviewed) study
that has appeared on medRxiv [52], did not find penetrat-
ing rare alleles associated with a severe disease phenotype
in four different cohorts analyzed by whole-exome or
whole-genome sequencing, thus questioning whether the
data by Zhang et al. [31] have general validity. These dis-
crepancies might be attributed at least in part to the het-
erogeneity of biobanks, to how phenotypic stratification is
clinically assessed, and to how functional studies are con-
ducted [31].
Identifying the role of rare variants is important in
order to improve predictive testing, to unravel the
pathogenetic mechanisms in different subgroups of
SARS-CoV-2 positive subjects, and to develop personal-
ized medicine for individual COVID-19 patients tailored
to his or her genetic background. It is possible that, in a
complex multifactorial and multigenic disease, such as
COVID-19, several genetic and epigenetic factors are
modulating the phenotypic manifestation, thus compli-
cating the analysis of genotype-phenotype correlations.
For example, it is known that non-coding RNAs
Novelli et al. Human Genomics (2021) 15:27 Page 4 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
(ncRNAs), and in particular microRNAs (miRNAs), are
involved in the pathogenesis of SARS-CoV-2 infection
and in host antiviral immune defense mechanisms [53].
Genes encoding miRNAs, like other genes, show inter-
individual genetic variability, and several studies have
shown that genetic variants in miRNA genes can, in
some cases, affect their expression, maturation, and even
affinity for their target genes [54,55]. Thus, the high
clinical variability of COVID-19 might be influenced by
polymorphisms in microRNA target sites (MTS) or in
miRNA sequences [55]. Genetic and epigenetic differ-
ences in miRNA expression in cells targeted by the virus
during entry could affect the effectiveness of antiviral re-
sponses and therefore disease severity. Interestingly, it
has recently been shown that level of expression of genes
encoding proteins involved in virus attachment and
entry (e.g., ACE2,TMPRSS2) varies with age and may
provide a biological rationale for variability in
Table 2 Genetic risk factors for severe COVID-19
SARS-CoV-2 susceptibility gene variant or haplotype Risk estimated
[OR]
Frequency
[MAF]
References
TLR3,UNC93B1,TICAM1,TBK1,IRF3,IRF7,IFNAR1,IFNAR2 (autosomal-
dominant model)
9 <0.001 Zhang et al. [31]
IRF7,IFNAR1 (autosomal-recessive model) >50 <0.001 Zhang et al. [31]
rs769208985missense variant of FURIN N.A. <0.001 Latini et al. [32]
rs150892504missense variant of ERAP2 N.A. 0.002 Hu et al. [33]
rs138763430missense variant of BRF2 N.A. 0.002 Hu et al. [33]
rs147149459missense variant of ALOXE3 N.A. 0.002 Hu et al. [33]
rs117665206missense variant of TMEM181 N.A. 0.006 Hu et al. [33]
rs114363287missense variant of TMPRSS2 N.A. 0.006 Latini et al. [32]
HLA DRB*27:07 N.A. 0.02 Novelli et al. [34]
rs749566153UTR variant of TYK2 1.6 0.03 Pairo-Castineira et al. [35]
rs73064425intronic variant of LZTFL1 2.1 0.08 Pairo-Castineira et al. [35], Ellinghaus
et al. [36]
rs11385942intronic variant of LZTFL1 1.8 0.07 Ellinghaus et al [36]
HLA DQB1*06:02 N.A. 0.08 Novelli et al. [34]
rs143334143intronic variant of CCHCR1 1.9 0.09 Pairo-Castineira et al. [35]
HLA DRB1*15:01 N.A. 0.10 Novelli et al. [34]
rs65980455UTR variant of IFITM3 N.A. 0.19 Kim et al. [37]
rs429358missense variant of APOE 2.3-2.4 0.20 Kuo et al. [38]
rs93801423UTR variant of HLA-G 13 0.29 Pairo-Castineira et al. [35]
rs2109069intronic variant of DPP9 1.4 0.33 Pairo-Castineira et al. [35]
rs75603675missense variant of TMPRSS2 N.A. 0.36 Latini et al. [32]
rs12329760missense variant of TMPRSS2 0.9 0.39 Hou et al. [39]
rs657152intronic variant of ABO 1.3 0.41 Ellinghaus et al. [36]
rs6020298intronic variant of TMEM189-UBE2V1 1.2 0.58 Wang et al. [40]
rs10735079intronic variant of OAS1/3 1.3 0.64 Pairo-Castineira et al. [35]
rs2236757intronic variant of IFNAR2 1.3 0.71 Pairo-Castineira et al. [35]
rs3131294intronic variant of NOTCH4 1.5 0.9 Pairo-Castineira et al. [35]
HLA B*46:01 2.1 N.A. Lin et al. [41]
HLA-E*0101/0103 2.1/2.7 N.A. Vietzen et al. [42]
KLRC2
del
2.6/7.1 N.A. Vietzen et al. [42]
HLA B*54:01 5.4 N.A. Lin et al. [41]
c.2129_2132del, p.Gln710Argfs*18frameshift variant of TLR7 N.A. N.A. van der Made et al. [43]
c.2383G>T, p.Val795Phemissense variant of TLR7 N.A. N.A. van der Made et al. [43]
rs140312271missense variant of ACE2 N.A. N.A. Novelli et al. [44]
MAF major allele frequency, N.A. not applicable, OR odds ratio
Novelli et al. Human Genomics (2021) 15:27 Page 5 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
presentation of COVID-19 [56]. Recently, Blume et al.
[57] identified a new short isoform of ACE2 expressed
in the airway epithelium. Short ACE2 is upregulated in
response to interferon stimulation and rhinovirus, but
not SARS-CoV-2 infection. Its expression is regulated
independently of the primary transcript, with putative
promoter elements identified upstream of the transcrip-
tional initiation site of the short ACE2 transcript. The
characterization of the functional elements of the ACE2
promoter and above all the factors involved in its regula-
tion will help to understand better the mechanisms of
pathology caused by SARS-CoV-2.
Newly the CHGE Consortium (Covid Human Genetic
Effort, https://www.covidhge.com/about) initiated a
study to enroll individuals (referred to as resistant)
who were not infected with SARS-CoV-2 despite re-
peated exposure (e.g., care-givers or familiars of a patient
with severe pneumonia), as evidenced by the absence of
the disease and virus specific antibody titers in several
tests. It is conceivable that these subjects carry mono-
genic variations that make them naturally resistant to
virus entry, as previously shown for the DARC gene and
Plasmodium vivax,CCR5 and HIV, and FUT2 and Nor-
ovirus [5860]. Currently, there are no publications on
this resistant cluster,but recently, Zeberg and Pääbo
[61] have identified an haplotype on chromosome 12,
which is associated with a 22% reduction in relative risk
of becoming severely ill with COVID-19 when infected
by SARS-CoV-2. Interestingly, this haplotype is inherited
from Neanderthals and it is present at substantial fre-
quencies in all regions of the world outside Africa. The
genomic region where this haplotype occurs encodes
proteins that are important during infections with RNA
viruses.
COVID-19 in Africa and in Latin America
Considering the multifactorial complexity of this disease,
its rapid epidemic spread, and its fast evolving causative
agent, COVID-19 is ravaging emerging countries, and its
impacts in low-resource areas deserves our attention
and action. We focus here on two regions of particular
interest: Africa and Latin America.
On January 1, 2021, J. M. Maeda and J. N. Nkenga-
song published a paper on The puzzle of the
COVID-19 pandemic in Africa[62]. The figure in
the paper depicts the two COVID-19 wavesseen in
Europe are much less evident in Africa: rather, there
has been a peak in July, 2020, followed by a gradual
decrease and in turn by a gradual increase that is still
on-going. The real puzzleis that a previous report
(issued on March 26, 2020, by the WHO Collaborat-
ing Centre for Infectious Disease Modeling; MRC
Centre for Global Infectious Disease) had predicted
some 70 million cases and some 3 million deaths in
Africa; in contrast, by November 22, 2020, the official
figures were 2,070,953 cases and 49,728 deaths. The
authors (who are at the Africa Centers for Disease
Control and Preventionthe African CDCin Addis
Ababa, Ethiopia) mention challenges with testingas
one possible reason for this striking discrepancy. This
notion is also supported by a compilation from
Nigeria [63]; according to official data, three-quarters
of African COVID-19 cases are in South Africa and
Egypt. An extreme case is that of Tanzania, where
the COVID-19 epidemic is officially finished, and the
last recorded case was on May 19, 2020. We think
limited testing is a major reason for the paucity of
cases in Africa.
Be that as it may, at least three factors of biological
and epidemiological interest may have contributed to
reducing the impact of the pandemic in Africa. First,
in tropical Africa, sun exposure and UV radiation
may inactivate an RNA virus rather more quickly.
However, this hypothesis is not confirmed in other
countries such as northern Brazil and other regions
ofthetropicswhereCOVID-19isfound.Further-
more, the mode of transmission that occurs through
droplets and aerosols does not fit this hypothesis.
Second, the age distribution of the population is
much younger. Third, a significant fraction of people
(perhaps up to 30%) [64] may have antibodies against
other corona viruses that cross-react with SARS-CoV-
2 (even though only 2-8% have specific anti-SARS-
CoV2 antibodies suggesting there may be, like in Eur-
ope, a large number of asymptomatic infections) [65].
Latin America has also been ravaged by COVID-19
and may be the worlds worst affected region [66].
Vaccinations against the virus are seen as the way
forward and have begun (https://www.as-coa.org/
articles/timeline-latin-americas-race-covid-19-vaccine)
[67]. We must support a path to a world that defeats
COVID-19 in all parts of the world. In fact inter-
national efforts by the Global Vaccine Alliance
(GAVI), Coalition for Epidemic Preparedness Innova-
tions (CEPI), Gates Foundations, WHO, etc. (https://
www.weforum.org/agenda/2020/06/vaccines-
immunization-poor-countries-coronavirus-covid-gavi)
[68] are thankfully ramping up.
New technologies accelerated into clinical
practice
The pandemic has produced an unprecedented shift
in the direction of basic and clinical research. We
think it is remarkable that so many highly qualified
research laboratories have been eager or willing to
bring their experience in other areas to bear on this
impelling worldwide problem, and we think it is com-
mendable that they have had the courage to literally
Novelli et al. Human Genomics (2021) 15:27 Page 6 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
re-purpose their work. There has been also a deluge
of publications: at March 30, 2021, the search item
COVID-19 in PubMed yields 118,065 papers; by the
time this paper is published, there will be several
thousands more publications. At the same time, new
technologies for disease treatment and prevention are
also being developed at unprecedented speed. In gen-
eral, it takes a long time for advances to reach con-
sumers, especially in the areas of biotechnology and
health. However, the pandemic has led to a dramatic
acceleration, including in the development of COVID-
19 vaccines (Table 3)[69].
Monoclonal antibodies
Although there are currently numerous studies to iden-
tify antivirals for SARS-CoV-2, the only compounds with
therapeutic efficacy already in use are monoclonal anti-
bodies (mAbs). mAbs are directed against the binding
site of the SARS-CoV-2 spike protein receptor and they
are able to block virus entry into human cells. mAbs
Table 3 Overview of the worldwide approved different types of COVID-19 vaccines
Vaccine Product
name
Vaccine
type
Phase
III
efficacy
Doses Storage Prize
per dose
Distribution
BioNTech/Pfizer BNT162b2 mRNA 95% 2 doses (0.3
mL) [21
days apart]
80 and 60 °C
(112 and 76
°F) for 6
months
+2 and +8 °C
(+36 and +46
°F) for 5 days
19.5 $ USA, EU, UK, Argentina, Australia, Bahrain,
Canada, Chile, Costa Rica, Ecuador, Hong Kong,
Iraq, Israel, Jordan, Kuwait, Malaysia, Mexico,
Oman, Panama, Philippines, Qatar, Saudi Arabia,
Singapore, South Korea, United Arab Emirates
COVAX (COVID-19 Vaccines Global Access)
Moderna mRNA-1273 mRNA 94.1% 2 doses (0.5
mL) [28
days apart]
20 °C (4 °F)
for 4 months
+2 and +8 °C
(+36 and +46
°F) for 30 days
32-37 $ USA, EU, UK, Canada, Greenland, Iceland, Israel,
Saudi Arabia, Singapore, Vietnam
COVAX (COVID-19 Vaccines Global Access)
Oxford/
AstraZeneca
ChAdOx1
AZD1222/
Vaxzevria
Viral vector 81.3% 2 doses (0.5
mL) [10-12
weeks
apart]
+2 and +8 °C
(+36 and +46
°F)
1.5-4 $ USA, EU, UK, Australia, Bangladesh, Brazil,
Canada, Greenland, India, Mexico, Nepal,
Pakistan, Philippines, Sri Lanka, Taiwan, Vietnam
COVAX (COVID-19 Vaccines Global Access)
Gamaleya
(Sputnik V)
Sputnik V
Gam-Covid-
Vac
Viral vector 91.6% 2 doses (0.5
mL)
[28 days
apart]
18 °C (0 °F)
for 24 months
+2 and +8 °C
(+36 and +46
°F) for 3
months
< 10 $ Russia, India, Brazil, China, South Korea, Hungary,
Argentina
Johnson&Johnson JNJ-
78436735
Ad26.COV2.S
Viral vector 72% 1 dose 18 °C (0 °F)
for 24 months
+2 and +8 °C
(+36 and +46
°F) for 3
months
10 $ USA, EU, Greenland, Canada
COVAX (COVID-19 Vaccines Global Access)
Novavax NVX-
CoV2373
Virus-like
particle
89% 2 doses [21
days apart]
18 °C (0 °F)
for 24 months
+2 and +8 °C
(+36 and +46
°F) for 3
months
16 $ USA, Canada
Sinopharm BBIBP-CorV Inactivated
virus
78% 2 doses [3
weeks
apart]
+2 and +8 °C
(+36 and
+46°F)
Three years in
storage
< 77 $ China, United Arab Emirates, Argentina, Bahrain,
Egypt, Marocco, Pakistan, Perù
SinoVac CoronaVac Inactivated
virus
50% 2 doses [3
weeks
apart]
2-8 °C (36-46
°F)
Three years in
storage
14 $ China, Brazil, Turkey, Chile, Indonesia, Philippines
Covaxin (Bharat
Biontech)
BBV152 Inactivated
virus
81% 2 doses [21
days apart]
2-8 °C (36-46
°F)
Three years in
storage
1 $ India, Iran, Mauritius, Nepal, Zimbabwe
Novelli et al. Human Genomics (2021) 15:27 Page 7 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
have this name because they are produced by one type
of immune cells (plasma cells) that are the progeny of a
single parent cell [70]. mAbs, are laboratory-produced
macromolecules engineered to bind to antigens of se-
lected targets (e.g., cancer cells, microorganisms, viruses)
[71]. The efficacy of mAbs has been successfully tested
in other coronavirus infections (SARS-CoV) [7274] and
MERS [75]. During the past 12 months, several SARS-
CoV-2 neutralizing mAbs have been isolated and charac-
terized in several clinical studies (NCT04452318,
NCT04497987). Some of these, such as bamlanivimab,
casirivimab, and imdevimab, have been approved for
emergency use in the treatment of mildly ill subjects.
mAbs regarded as good candidates for clinical use have
been derived by cloning B cells from patients who have
recovered from COVID-19, or from other natural
sources [7682]. Bamlanivimab has been associated with
a decrease in viral load and the frequency of hospitaliza-
tions or emergency department visits in outpatients with
COVID-19 [83]; however, when it was co-administered
with Remdesivir, it did not demonstrate efficacy among
hospitalized patients who had COVID-19 without end-
organ failure [84]. In a double-blind, phase I-III trial in-
volving non-hospitalized COVID-19 patients, Weinreich
et al. [85] investigated two fully neutralizing mAbs used
in combination (casirivimab/imdevimab or REGN-
COV2), in the aim to reduce the risk that treatment-
resistant virus variants may emerge. REGN-COV2, de-
veloped by Regeneron Pharmaceuticals (USA), reduced
viral load, with a greater effect in patients whose im-
mune response to the virus had not yet been initiated, or
who had a high viral replication at baseline. Very recent
and unpublished data show that treatment with bamlani-
vimab (LY-CoV555) and etesevimab (LY-CoV016) to-
gether reduced the risk of hospitalization and death
from COVID-19 by 70% (https://investor.lilly.com/news-
releases/news-release-details/new-data-show-treatment-
lillys-neutralizing-antibodies)[86]. Recently, EMA (Euro-
pean Medicines Agency) has completed review on the
use of an additional mAb, regdanvimab (also known as
CT-P59) to treat COVID-19 patients and concluded that
regdanvimab can be used for the treatment of confirmed
COVID-19 in adult patients who do not require supple-
mental oxygen therapy and who are at high risk of pro-
gressing to severe outcome (https://www.ema.europa.eu/
en/news/ema-review-regdanvimab-covid-19-support-
national-decisions-early-use)[87].
mAbs may be also useful for the prophylaxis of
COVID-19 in persons who are at high-risk (health
workers and first responders, pregnant women, ring-
vaccination-type response to disease outbreak). By apply-
ing an innovative strategy, Miersch et al. [88,89] have
isolated high affinity synthetic mAbs from a phage li-
brary and thus developed a dynamic and rapid
technological platform; this approach has the potential
to identify in a short time mAbs against new virus vari-
ants. Synthetic engineering technologies may prove su-
perior to natural cloning methods, as they offer exquisite
control over the design of mAbs, that can prove more
efficient. Similarly, Rappazzo et al. [90] identified rare
broadly neutralizing antibodies (bnAbs) which can be
engineered for improve neutralization potency and pro-
tection in vivo. Synthetic methods have the added ad-
vantage that they do not depend on natural repertoires,
i.e., they are not limited by the need of accessing in-
fected patients as a source of therapeutic agents.
Andreano et al. [91] have isolated and characterized ex-
tremely potent neutralizing mAbs, suitable for prophy-
lactic and therapeutic interventions of wild-type SARS-
CoV-2 as well as emerging variants. Remarkably, an
international research consortium recently developed a
bispecific monoclonal antibody targeting two different
SARS-CoV-2 sites, thereby preventing the virus from
mutating to resist therapy. A single injection of the bis-
pecific antibody provided protection against disease in
mice. The antibody effectively reduced the viral load in
the lungs and mitigated the typical COVID-19 inflam-
mation [92].
mRNA vaccines
Vaccines against SARS-CoV-2 infection were made in
less than a year (Table 3). This research has undoubtedly
been a great success of modern technology and public
and private investments that have allowed to accelerate
many of the processes required to develop a vaccine.
But, of course, mRNA vaccines represent an epochal sci-
entific and technical breakthrough.
mRNA was discovered in 1961 by Brenner et al. [93]
and its ability to form drugs has been described already
in 1989 [94,95]. Since then, dozens of studies on the
subject have been published, including a study on a vac-
cine against the MERS-CoV, which allowed the acceler-
ated development of the SARS-CoV-2 vaccine [96]. This
technology enables the synthetic production of the
mRNA encoding any protein. mRNA vaccines have ad-
vantages over DNA vaccines. In general, lower doses are
required to induce an immune response. Synthetic
mRNA does not integrate into the cells genome, and no
transcriptional step is involved, as the mRNA is directly
translated, and then it undergoes degradation [97]. The
vaccine activates both humoral immune system and cel-
lular immune response, similar to live virus attenuated
vaccine [98100].
ThesafetydatainadultsonmRNAvaccinesagainst
COVID-19 have been reassuring. The most common side
effects in adults are only local, within 7 days of receiving
the first inoculation, and more common after the second
dose [101,102]. The side effects are mild-to-moderate; no
Novelli et al. Human Genomics (2021) 15:27 Page 8 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
severe-grade local side effects were reported [98,103]. Sys-
temic signs (fever of 38 °C) were reported in 75% of vac-
cine recipients. Subjects over the age of 65 suffered less
systemic symptoms, but reported fatigue and headaches.
During the rollout, some severe allergic reactions were
also observed, but they were rare [104].
RNA-based vaccines efficacy, safety, and the potential
for rapid, inexpensive, and scalable production makes
them a powerful advantageous option to combat
COVID-19, including probably new variants [105110].
Widge et al. [111] have reported immunogenicity data
119 days after the first dose of vaccine (i.e., 90 days after
the second dose) in 34 healthy adult participants who re-
ceived two injections of 100 μg each. They provided fur-
ther evidence that mRNA-1273, developed by National
Institute of Allergy and Infectious Diseases (NIAID), the
Biomedical Advanced Research and Development Au-
thority (BARDA), and Moderna (ModeRNA Therapeu-
tics, USA) have the potential to provide a durable
humoral immunity, although for how long is of course
not yet known.
Recently, a CDC study of about 4000 healthcare
workers and workers belonging to essential categories
were tested weekly for coronavirus after administration
of the mRNA vaccine. The analyses found that people
who had completed the vaccination course had a 90%
reduced chance of becoming infected. Furthermore, the
study showed that already from the first dose, the per-
centage after 14 days from the first injection was already
close to 80%. Results clearly indicate the vaccines ability
to interrupt viral transmission [112].
mRNA-based drugs are a promising novel platform
that might be useful for the development of vaccines
against emerging pandemic infectious diseases [113].
Furthermore, unconventional tools such as Cas13a-
crRNA complex, an RNA-activated RNase, are being ex-
plored as novel therapeutics against SARS-CoV-2 infec-
tion, displaying promising results in reducing viral
replication and symptoms in animal models [114]. RNA
in human cells is susceptible to editing, and the RNA
genome of SARS-CoV-2 is not an exemption. From a
comprehensive sequence analysis of Coronaviridae, nu-
cleotide changes have been identified that may be signa-
tures of RNA editing by both ADAR deaminases and
APOBEC deaminases. It has been suggested that this
process may contribute to shaping the fate of both virus
and patient [115].
Conclusions
We are heartened by the progress in therapeutics and
preventive strategies for COVID-19. Having achieved
within less than 1 year from the start of the pandemic
not only the development of a vaccine but also of several
vaccines; having conducted the necessary clinical trials;
having obtained approval from multiple regulatory agen-
cies around the world; having actually already carried
out mass vaccinations at least in some countries, is noth-
ing short of spectacular [68]. While we can certainly not
relent on the standard preventive measures (rigorous so-
cial distancing, frequent and thorough handwashing,
avoiding poorly ventilated spaces, wearing of face masks
as warranted), we are confident that the vaccines cur-
rently available and those in the pipeline will help defeat
COVID-19 and allow a return to normality in due
course [116,117]. Improved therapies, such as mAbs,
will also be useful in treating the disease. Understanding
the virus entry and egress mechanisms could also open
the door to promising therapeutic perspectives [118,
119]. Molecular testing and genomics will play a critical
role in the detection of newly emerging variants.
Furthermore, we are mindful of our fellow humans in
other countries, particularly in emerging economies. For
many years, the Global Fund has provided funding for
those affected by HIV; it has been a generous gesture,
initially fueled by concerns that from a reservoir in Af-
rica transmission elsewhere could take place. If HIV has
been an epidemic, SARS-CoV-2 has sparked a pandemic,
and it will be in everybodys interest that it is controlled
on a global scale because, as stated at the beginning of
this paper, otherwise it will continue to affect the world.
We are aware that WHO has launched the COVAX
(COVID-19 Vaccines Global Access) scheme, and that
the GAVI, backed by the Bill & Melinda Gates Founda-
tion, the WHO, the World Bank, UNICEF, and others,
has pledged $8.8 billion to reduce vaccine costs for poor
countries. One possibility is that a good part of these
funds will be absorbed by vaccinating persons from poor
countries and provide immunization certificates to those
who travel to richer countries, so that the latter will be
protected. But we call on international organizations to
set aside this potential narrow-sighted approach: and
make instead vaccines available to all nations on a wide
scale. We look forward to the WHO leading rigorously
and expeditiously, lest their effectiveness is brought into
question more than it has been hitherto. It is legitimate
to hope that the lessons learned with COVID-19 will
help in the future, not only in being more prepared for
other similar emergencies but also in acting as one, for
we have only one planet and only one humankind.
Abbreviations
ARDS: Acute respiratory distress syndrome; ASEAN: Organization of Southeast
Asian States; AU: African Union; BARDA: Biomedical Advanced Research and
Development Authority; CDC: Centers for Disease Control; CEPI: Coalition for
Epidemic Preparedness Innovations; CHGE: Covid Human Genetic Effort;
COPD: Chronic obstructive pulmonary disease; COVAX: COVID-19 Vaccines
Global Access; COVID-19: Coronavirus disease 2019; CTAP: Coronavirus
Treatment Acceleration Program; EMA: European Medicine Agency;
EU: European Union; EUA: Emergency Use Authorization; FAO: Food and
Agriculture Organization; FDA: Food and Drug Administration; GAVI: Global
Vaccine Alliance; GWAS: Genome Wide Association Studies; IFR: Infection
Novelli et al. Human Genomics (2021) 15:27 Page 9 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
fatality rate; mAbs: Monoclonal antibodies; miRNA: micro RNA; MERS: Middle
East respiratory syndrome; NIAID: National Institute of Allergy and Infectious
Diseases; OAS: Organization of American States; ORF: Open reading frame;
OSAS: Obstructive sleep apnea syndrome; SARS-CoV-2: Severe acute
respiratory syndrome coronavirus 2; UN: United Nations; WHO: World Health
Organization
Supplementary Information
The online version contains supplementary material available at https://doi.
org/10.1186/s40246-021-00326-3.
Additional file 1: Table S1. List of genomes used in the analysis. We
gratefully acknowledge the following Authors from the Originating
laboratories responsible for obtaining the specimens, as well as the
Submitting laboratories where the genome data were generated and
shared via GISAID, on which this research is based.
Acknowledgements
We are particularly grateful for the assistance given by Dr. Francesca Pisanu
for her continuous help with the editing and organization of the manuscript.
Authorscontributions
G.N. and J.K.V.R. conceived the paper, performed the systematic review,
wrote and edited the manuscript. R.M.S. wrote mRNA vaccines paragraph.
J.K.V.R. is also responsible for the sections on acceleration of technologies
and politics along with some revisions. V.L.C. conceived and edited Table 2,
Table 3, and revised the paper. A.F.B. generated Fig. 1. M.B., V.V., A.F.B., L.L.,
and N.D.G. performed review and revision of the paper. All authors read and
approved the final version of the manuscript.
Funding
This study was also supported in part by a grant of Regione Lazio (Italy,
Progetti di Gruppi di Ricerca 2020 A0375-2020-36663 GecoBiomark) and
Rome Foundation (Italy, Prot 317A/I) to GN and NIH N. AA028432 to V.V.
Availability of data and materials
Data sharing is not applicable to this article as no datasets were generated
or analyzed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Author details
1
Department of Biomedicine and Prevention, Tor VergataUniversity of
Rome, 00133 Rome, Italy.
2
IRCCS Neuromed, Pozzilli, IS, Italy.
3
Department of
Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557,
USA.
4
Department of Biology, Tor Vergata University of Rome, 00133 Rome,
Italy.
5
Pediatric Hemato-Oncology, Sheba Medical Center, Tel Hashomer,
Israel.
6
Department of Epidemiology of Microbial Diseases, Yale School of
Public Health, New Haven, CT 06510, USA.
7
Department of Environmental
Health Sciences, Yale School of Public Health, New Haven, CT 06510, USA.
8
Haematology, Muhimbili University of Health and Allied Sciences, Dar-es
Salaam, Tanzania.
9
Australian Institute of Tropical Health and Medicine, James
Cook University, Smithfield, Queensland 4878, Australia.
Received: 8 February 2021 Accepted: 15 April 2021
References
1. Green MS. Did the hesitancy in declaring COVID-19 a pandemic reflect a
need to redefine the term? Lancet. 2020;395(10229):10345. https://doi.
org/10.1016/S0140-6736(20)30630-9.
2. U.S. Food & Drug Administration (2021). Fda.gov fda.gov. Retrieved 31/03,
2021, from https://www.fda.gov/drugs/coronavirus-covid-19-drugs/corona
virus-treatment-acceleration-program-ctap.
3. Chaib, F. (2020). Independent evaluation of global COVID-19 response
announced. Retrieved 27/01, 2021, from https://www.who.int/news/item/
09-07-2020-independent-evaluation-of-global-covid-19-response-a
nnounced.
4. Kupferschmidt K, Vogel G (2021). Sciencemag.org sciencemag.org.
Retrieved 31/03, 2021, from https://www.sciencemag.org/news/2021/03/
compromise-who-report-resolves-little-pandemic-s-origins-details-probe-s-
next-steps.
5. Robson F, Khan KS, Le TK, Paris C, Demirbag S, Barfuss P, et al. Coronavirus
RNA proofreading: molecular basis and therapeutic targeting. Mol Cell.
2020;79(5):71027Erratum in: Mol Cell. 2020 Dec 17;80(6):1136-1138. https://
doi.org/10.1016/j.molcel.2020.11.048.
6. Duffy S, Shackelton LA, Holmes EC. Rates of evolutionary change in viruses:
patterns and determinants. Nat Rev Genet. 2008;9(4):26776. https://doi.
org/10.1038/nrg2323.
7. Daugherty MD, Malik HS. Rules of engagement: molecular insights from
host-virus arms races. Annu Rev Genet. 2012;46(1):677700. https://doi.org/1
0.1146/annurev-genet-110711-155522.
8. Mandary MB, Masomian M, Poh CL. Impact of RNA virus evolution on
quasispecies formation and virulence. Int J Mol Sci. 2019;20(18):4657. https://
doi.org/10.3390/ijms20184657.
9. Stern A, Bianco S, Yeh MT, Wright C, Butcher K, Tang C, et al. Costs and
benefits of mutational robustness in RNA viruses. Cell Rep. 2014;8(4):1026
36. https://doi.org/10.1016/j.celrep.2014.07.011.
10. GISAID (2020). Global initiative on sharing avian flu data. Retrieved 31/03,
2021, from https://www.gisaid.org/.
11. Nextstrain (2021). Real-time tracking of pathogen evolution. Retrieved 31/
03, 2021, from https://nextstrain.org/ncov/global.
12. Hadfield J, Megill C, Bell SM, Huddleston J, Potter B, Callender C, et al.
Nextstrain: real-time tracking of pathogen evolution. Bioinformatics. 2018;
34(23):41213. https://doi.org/10.1093/bioinformatics/bty407.
13. Huang SW, Miller SO, Yen CH, Wang SF. Impact of genetic variability in
ACE2 expression on the evolutionary dynamics of SARS-CoV-2 spike D614G
mutation. Genes (Basel). 2020;12(1):E16.
14. Wise J. Covid-19: new coronavirus variant is identified in UK. BMJ. 2020;371:
m4857.
15. Plante JA, Liu Y, Liu J, Xia H, Johnson BA, Lokugamage KG, et al. Spike
mutation D614G alters SARS-CoV-2 fitness. Nature. 2021; 592(7852):11621.
https://doi.org/10.1038/s41586-020-2895-3.
16. Kemp SA, Datir RP, Collier DA, Ferreira IATM, Carabelli A, Harvey W,
Robertson DL, Gupta RK. Recurrent emergence and transmission of a SARS-
CoV-2 Spike deletion ΔH69/ΔV70. bioRxiv. 2020. https://doi.org/10.1101/202
0.12.14.422555.
17. Tegally H, Wilkinson E, Giovanetti M, Iranzadeh A, Fonseca V, Giandhari J,
et al. Emergence and rapid spread of a new severe acute respiratory
syndrome-related coronavirus 2 (SARS-CoV-2) lineage with multiple spike
mutations in South Africa. medRxiv. 2020. https://doi.org/10.1101/202
0.12.21.20248640.
18. Da Silva FR Jr, Benites FJ, Lamarca AP, de Almeida LGP, Hansen AW, Gularte
JS, et al. Pervasive transmission of E484K and emergence of VUI-NPI3L with
evidence of SARS-CoV-2 co-infection events by two lineages in Rio Grande
do Sul, Brasil.Virus Res. 2021;296:198345.https://doi.org/10.1016/j.virusres.2
021.198345.
19. Deng X, Garcia-Knight MA, Khalid MM, Servellita V, Wang C, Morris MK, et al.
Transmission, infectivity, and antibody neutralization of an emerging SARS-
CoV-2 variant in California carrying a L452R spike protein mutation.
medRxiv. 2021. https://doi.org/10.1101/2021.03.07.21252647.
20. Abdool Karim SS, de Oliveira T. New SARS-CoV-2 variants - clinical, public
health, and vaccine implications. N Engl J Med. 2021. https://doi.org/10.1
056/NEJMc2100362.
Novelli et al. Human Genomics (2021) 15:27 Page 10 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
21. Tarke A, Sidney J, Methot N, Zhang Y, Dan JM, Goodwin B, et al. Negligible
impact of SARS-CoV-2 variants on CD4
+
and CD8
+
T cell reactivity in COVID-
19 exposed donors and vaccinees. bioRxiv. 2021. https://doi.org/10.1101/2
021.02.27.433180.
22. Challen R, Brooks-Pollock E, Read JM, Dyson L, Tsaneva-Atanasova K, Danon
L. Risk of mortality in patients infected with SARS-CoV-2 variant of concern
202012/1: matched cohort study. BMJ. 2021. https://doi.org/10.1136/bmj.
n579.
23. Alpert T, Lasek-Nesselquist E, Brito AF, Valesano AL, Rothman J, MacKay MJ,
et al. Early introductions and community transmission of SARS-CoV-2 variant
B.1.1.7 in the United States. medRxiv.2021.02.10.21251540.
24. Cyranoski D. Alarming COVID variants show vital role of genomic
surveillance. Nature. 2021;589(7842):3378. https://doi.org/10.1038/d41586-
021-00065-4.
25. Levin AT, Hanage WP, Owusu-Boaitey N, Cochran KB, Walsh SP, Meyerowitz-
Katz G. Assessing the age specificity of infection fatality rates for COVID-19:
systematic review, meta-analysis, and public policy implications. Eur J
Epidemiol. 2020;35(12):112338. https://doi.org/10.1007/s10654-020-00698-1.
26. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of
patients infected with 2019 novel coronavirus in Wuhan, China. Lancet.
2020;395(10223):497506. https://doi.org/10.1016/S0140-6736(20)30183-5.
27. Novelli L, Motta F, De Santis M, Ansari AA, Gershwin ME, Selmi C. The
JANUS of chronic inflammatory and autoimmune diseases onset during
COVID-19 - a systematic review of the literature. J Autoimmun. 2020;117:
102592.
28. Guler SA, Ebner L, Beigelman C, Bridevaux PO, Brutsche M, Clarenbach C,
et al. Pulmonary function and radiological features four months after
COVID-19: first results from the national prospective observational Swiss
COVID-19 lung study. Eur Respir J. 2021:2003690. https://doi.org/10.1183/13
993003.03690-2020.
29. Di Maria E, Latini A, Borgiani P, Novelli G. Genetic variants of the human
host influencing the coronavirus-associated phenotypes (SARS, MERS and
COVID-19): rapid systematic review and field synopsis. Hum Genomics. 2020;
14(1):30. https://doi.org/10.1186/s40246-020-00280-6.
30. Zhang Q, Bastard P, Bolze A, Jouanguy E, Zhang SY, Cobat A, et al. Life-
threatening COVID-19: defective interferons unleash excessive inflammation.
Med. 2020;1(1):1420.
31. Zhang Q, Bastard P, Liu Z, Le Pen J, Moncada-Velez M, Chen J, et al. Inborn
errors of type I IFN immunity in patients with life-threatening COVID-19.
Science. 2020;370(6515):eabd4570. https://doi.org/10.1126/science.abd45702
020:eabd4570.
32. Latini A, Agolini E, Novelli A, Borgiani P, Giannini R, Gravina P, et al. COVID-
19 and genetic variants of protein involved in the SARS-CoV-2 entry into
the host cells. Genes (Basel). 2020;11(9):1010. https://doi.org/10.3390/
genes11091010.
33. Hu J, Li C, Wang S, Li T, Zhang H. Genetic variants are identified to increase
risk of COVID-19 related mortality from UK Biobank data. medRxiv. 2020.11.
05.20226761.
34. Novelli A, Andreani M, Biancolella M, Liberatoscioli L, Passarelli C,
Colona VL, et al. HLA allele frequencies and susceptibility to COVID-19
in a group of 99 Italian patients. HLA. 2020;96(5):6104. https://doi.org/1
0.1111/tan.14047.
35. Pairo-Castineira E, Clohisey S, Klaric L, Bretherick AD, Rawlik K, Pasko D, et al.
Genetic mechanisms of critical illness in Covid-19. Nature. 2020.
36. Severe Covid-19 GWAS Group, Ellinghaus D, Degenhardt F, Bujanda L, Buti
M, Albillos A, et al. Genomewide association study of severe Covid-19 with
respiratory failure. N Engl J Med. 2020;383(16):152234. https://doi.org/10.1
056/NEJMoa2020283.
37. Kim YC, Jeong BH. Strong correlation between the case fatality rate of
COVID-19 and the rs6598045 single nucleotide polymorphism (SNP) of the
interferon-induced transmembrane protein 3 (IFITM3) gene at the
population-level. Genes (Basel). 2020;12(1):42. https://doi.org/10.3390/
genes12010042.
38. Kuo CL, Pilling LC, Atkins JL, Masoli JAH, Delgado J, Kuchel GA, et al. APOE
e4 genotype predicts severe COVID-19 in the UK biobank community
cohort. J Gerontol A Biol Sci Med Sci. 2020;75(11):22312. https://doi.org/1
0.1093/gerona/glaa131.
39. Hou Y, Zhao J, Martin W, Kallianpur A, Chung MK, Jehi L, et al. New insights
into genetic susceptibility of COVID-19: an ACE2 and TMPRSS2
polymorphism analysis. BMC Med. 2020;18(1):216. https://doi.org/10.1186/
s12916-020-01673-z.
40. Wang F, Huang S, Gao R, Zhou Y, Lai C, Li Z, et al. Initial whole-genome
sequencing and analysis of the host genetic contribution to COVID-19
severity and susceptibility. Cell Discov. 2020;6(1):83. https://doi.org/10.1038/
s41421-020-00231-4.
41. Lin M, Tseng HK, Trejaut JA, Lee HL, Loo JH, Chu CC, et al. Association of
HLA class I with severe acute respiratory syndrome coronavirus infection.
BMC Med Genet. 2003;4(1):9. https://doi.org/10.1186/1471-2350-4-9.
42. Vietzen H, Zoufaly A, Traugott M, Aberle J, Aberle SW, Puchhammer-Stöckl
E. Deletion of the NKG2C receptor encoding KLRC2 gene and HLA-E
variants are risk factors for severe COVID-19. Genet Med. 2021:15.
43. van der Made CI, Simons A, Schuurs-Hoeijmakers J, van den Heuvel G,
Mantere T, Kersten S, et al. Presence of genetic variants among young men
with severe COVID-19. JAMA. 2020;324(7):111. https://doi.org/10.1001/ja
ma.2020.13719.
44. Novelli A, Biancolella M, Borgiani P, Cocciadiferro D, Colona VL, DApice MR,
et al. Analysis of ACE2 genetic variants in 131 Italian SARS-CoV-2-positive
patients. Hum Genomics. 2020;14(1):29. https://doi.org/10.1186/s40246-020-
00279-z.
45. Novelli G, Biancolella M, Mehrian-Shai R, Erickson C, Godri Pollitt KJ, Vasiliou
V, et al. COVID-19 update: the first 6 months of the pandemic. Human
Genom. 2020;14(1):48. https://doi.org/10.1186/s40246-020-00298-w.
46. Curtis D. Variants in ACE2 and TMPRSS2 genes are not major determinants
of COVID-19 severity in UK Biobank subjects. medRxiv. 2020:2020.05.01.
20085860.
47. Elhabyan A, Elyaacoub S, Sanad E, Abukhadra A, Elhabyan A, Dinu V. The
role of host genetics in susceptibility to severe viral infections in humans
and insights into host genetics of severe COVID-19: a systematic review.
Virus Res. 2020;289:198163. https://doi.org/10.1016/j.virusres.2020.198163.
48. Oh JH, Tannenbaum A, Deasy JO. Identification of biological correlates associated
with respiratory failure in COVID-19. BMC Med Genet. 2020;13(1):186.
49. Yuan J, Fan D, Xue Z, Qu J, Su J. Co-expression of mitochondrial genes and
ACE2 in cornea involved in COVID-19. Invest Ophthalmol Vis Sci. 2020;
61(12):13. https://doi.org/10.1167/iovs.61.12.13.
50. Smatti MK, Al-Sarraj YA, Albagha O, Yassine HM. Host genetic variants
potentially associated with SARS-CoV-2: a multi-population analysis. Front
Genet. 2020;11(1064).
51. Duncan L, Shen H, Gelaye B, Meijsen J, Ressler K, Feldman M, et al. Analysis
of polygenic risk score usage and performance in diverse human
populations. Nat Commun. 2019;10(1):3328. https://doi.org/10.1038/s41467-
019-11112-0.
52. Povysil G, Butler-Laporte G, Shang N, Weng C, Khan A, Alaamery M, et al.
Failure to replicate the association of rare loss-of-function variants in type I
IFN immunity genes with severe COVID-19. medRxiv. 2020:2020.12.18.
20248226.
53. Henzinger H, Barth DA, Klec C, Pichler M. Non-coding RNAs and SARS-
related coronaviruses. Viruses. 2020;12(12).
54. Liu X, Han Z, Yang C. Associations of microRNA single nucleotide
polymorphisms and disease risk and pathophysiology. Clin Genet. 2017;
92(3):23542. https://doi.org/10.1111/cge.12950.
55. Moszyńska A, Gebert M, Collawn JF, Bartoszewski R. SNPs in microRNA
target sites and their potential role in human disease. Open Biol. 2017;7(4).
56. Schuler BA, Habermann AC, Plosa EJ, Taylor CJ, Jetter C, Negretti NM, et al.
Age-determined expression of priming protease TMPRSS2 and localization
of SARS-CoV-2 in lung epithelium. J Clin Invest. 2021;131(1). https://doi.org/1
0.1172/JCI140766.
57. Blume C, Jackson CL, Spalluto CM, Legebeke J, Nazlamova L, Conforti F, et al. A
novel ACE2 isoform is expressed in human respiratory epithelia and is
upregulated in response to interferons and RNA respiratory virus infection. Nat
Genet. 2021;53(2):20514. https://doi.org/10.1038/s41588-020-00759-x.
58. Miller LH, Mason SJ, Clyde DF, McGinniss MH. The resistance factor to
plasmodium vivax in blacks. The Duffy-blood-group genotype, FyFy. N Engl
J Med. 1976;295(6):3024. https://doi.org/10.1056/NEJM197608052950602.
59. Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, et al.
Genetic restriction of HIV-1 infection and progression to AIDS by a deletion
allele of the CKR5 structural gene. Hemophilia growth and development
study, multicenter AIDS cohort study, multicenter hemophilia cohort study,
san Francisco City cohort, ALIVE study. Science. 1996;273(5283):185662.
https://doi.org/10.1126/science.273.5283.1856.
60. Lindesmith L, Moe C, Marionneau S, Ruvoen N, Jiang X, Lindblad L, et al.
Human susceptibility and resistance to Norwalk virus infection. Nat Med.
2003;9(5):54853. https://doi.org/10.1038/nm860.
Novelli et al. Human Genomics (2021) 15:27 Page 11 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
61. Zeberg H, Pääbo S. A genomic region associated with protection against
severe COVID-19 is inherited from Neandertals. Proc Natl Acad Sci. 2021;
118(9):e2026309118. https://doi.org/10.1073/pnas.2026309118.
62. Maeda JM, Nkengasong JN. The puzzle of the COVID-19 pandemic in Africa.
Science. 2021;371(6524):278. https://doi.org/10.1126/science.abf8832.
63. Aluko OM, Lawal SA, Falana MM, Adeagbo AS, Ijomone OM. Tackling
COVID-19 in Africa: a focus on Nigerias peculiarities and challenges.
Innovation (N Y). 2021;2(1):100078. https://doi.org/10.1016/j.xinn.2021.1
00078.
64. Tso FY, Lidenge SJ, Peña PB, Clegg AA, Ngowi JR, Mwaiselage J, et al. High
prevalence of pre-existing serological cross-reactivity against severe acute
respiratory syndrome coronavirus-2 (SARS-CoV-2) in sub-Saharan Africa. Int J
Infect Dis. 2021;102:57783. https://doi.org/10.1016/j.ijid.2020.10.104.
65. Uyoga S, Adetifa IMO, Karanja HK, Nyagwange J, Tuju J, Wanjiku P,
et al. Seroprevalence of anti-SARS-CoV-2 IgG antibodies in Kenyan
blood donors. Science. 2021;371(6524):7982. https://doi.org/10.1126/
science.abe1916.
66. The Lancet. COVID-19 in Latin America: a humanitarian crisis. Lancet. 2020;
396(10261):1463. https://doi.org/10.1016/S0140-6736(20)32328-X.
67. Horwitz, L. (2021). Timeline: Latin Americas race for a COVID-19 vaccine.
Retrieved 31/03, 2021, from https://www.as-coa.org/articles/timeline-latin-a
mericas-race-covid-19-vaccine.
68. Kelland, K. (2021). COVID-19: Global donors pledge $8.8 billion for vaccines.
Retrieved 31/03, 2021, from https://www.weforum.org/agenda/2020/06/va
ccines-immunisation-poor-countries-coronavirus-covid-gavi.
69. Cohen J. Shots of hope. Science. 2020;370(6523):13924. https://doi.org/1
0.1126/science.370.6523.1392.
70. Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and
inflammation. Nat Rev Immunol. 2010;10(5):30116. https://doi.org/10.1038/
nri2761.
71. Casadevall A, Dadachova E, Pirofski LA. Passive antibody therapy for
infectious diseases. Nat Rev Microbiol. 2004;2(9):695703. https://doi.org/10.1
038/nrmicro974.
72. ter Meulen J, Bakker AB, van den Brink EN, Weverling GJ, Martina BE,
Haagmans BL, et al. Human monoclonal antibody as prophylaxis for SARS
coronavirus infection in ferrets. Lancet. 2004;363(9427):213941. https://doi.
org/10.1016/S0140-6736(04)16506-9.
73. Sui J, Li W, Murakami A, Tamin A, Matthews LJ, Wong SK, et al. Potent
neutralization of severe acute respiratory syndrome (SARS) coronavirus by a
human mAb to S1 protein that blocks receptor association. Proc Natl Acad
Sci U S A. 2004;101(8):253641. https://doi.org/10.1073/pnas.0307140101.
74. Zhu Z, Chakraborti S, He Y, Roberts A, Sheahan T, Xiao X, et al. Potent cross-
reactive neutralization of SARS coronavirus isolates by human monoclonal
antibodies. Proc Natl Acad Sci U S A. 2007;104(29):121238. https://doi.org/1
0.1073/pnas.0701000104.
75. Corti D, Zhao J, Pedotti M, Simonelli L, Agnihothram S, Fett C, et al.
Prophylactic and postexposure efficacy of a potent human monoclonal
antibody against MERS coronavirus. Proc Natl Acad Sci U S A. 2015;112(33):
104738. https://doi.org/10.1073/pnas.1510199112.
76. CaoY,SuB,GuoX,SunW,DengY,BaoL,etal.Potentneutralizing
antibodies against SARS-CoV-2 identified by high-throughput single-
cell sequencing of convalescent patientsB cells. Cell. 2020;182(1):73
84.e16.
77. Hansen J, Baum A, Pascal KE, Russo V, Giordano S, Wloga E, et al. Studies in
humanized mice and convalescent humans yield a SARS-CoV-2 antibody
cocktail. Science. 2020;369(6506):10104. https://doi.org/10.1126/science.a
bd0827.
78. Noy-Porat T, Makdasi E, Alcalay R, Mechaly A, Levy Y, Bercovich-Kinori A,
et al. A panel of human neutralizing mAbs targeting SARS-CoV-2 spike at
multiple epitopes. Nat Commun. 2020;11(1):4303. https://doi.org/10.1038/
s41467-020-18159-4.
79. Rogers TF, Zhao F, Huang D, Beutler N, Burns A, He WT, et al. Isolation of
potent SARS-CoV-2 neutralizing antibodies and protection from disease in a
small animal model. Science. 2020;369(6506):95663. https://doi.org/10.112
6/science.abc7520.
80. Shi R, Shan C, Duan X, Chen Z, Liu P, Song J, et al. A human neutralizing
antibody targets the receptor-binding site of SARS-CoV-2. Nature. 2020;
584(7819):1204. https://doi.org/10.1038/s41586-020-2381-y.
81. Wan J, Xing S, Ding L, Wang Y, Gu C, Wu Y, et al. Human-IgG-neutralizing
monoclonal antibodies block the SARS-CoV-2 infection. Cell Rep. 2020;32(3):
107918. https://doi.org/10.1016/j.celrep.2020.107918.
82. Wec AZ, Wrapp D, Herbert AS, Maurer DP, Haslwanter D, Sakharkar M, et al.
Broad neutralization of SARS-related viruses by human monoclonal antibodies.
Science. 2020;369(6504):7316. https://doi.org/10.1126/science.abc7424.
83. Chen P, Nirula A, Heller B, Gottlieb RL, Boscia J, Morris J, et al. SARS-CoV-2
neutralizing antibody LY-CoV555 in outpatients with Covid-19. N Engl J
Med. 2020.
84. Lundgren JD, Grund B, Barkauskas CE, Holland TL, Gottlieb RL, Sandkovsky
U, et al. A neutralizing monoclonal antibody for hospitalized patients with
Covid-19. N Engl J Med. 2020;384(10):90514. https://doi.org/10.1056/
NEJMoa2033130.
85. Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, et al.
REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid-19.
N Engl J Med. 2020;384(3):23851.https://doi.org/10.1056/NEJMoa2035002.
86. Lilly.com (2021). New data show treatment with Lillys neutralizing
antibodies bamlanivimab (LY-CoV555) and etesevimab (LY-CoV016)
together reduced risk of COVID-19 hospitalizations and death by 70
percent. Retrieved 31/03, 2021, from https://investor.lilly.com/news-relea
ses/news-release-details/new-data-show-treatment-lillys-neutralizing-a
ntibodies.
87. European Medicines Agency (EMA) (2021). Ema.europa.eu ema.europa.ue.
Retrieved 31/03, 2021 from https://www.ema.europa.eu/en/news/ema-
review-regdanvimab-covid-19-support-national-decisions-early-use.
88. Miersch S, Ustav M, Li Z, Case JB, Ganaie S, Matusali G, et al. Synthetic
antibodies neutralize SARS-CoV-2 infection of mammalian cells. bioRxiv.
2020:2020.06.05.137349.
89. Miersch S, Li Z, Saberianfar R, Ustav M, Case JB, Blazer L, et al. Tetravalent
SARS-CoV-2 neutralizing antibodies show enhanced potency and resistance
to escape mutations. bioRxiv. 2020.
90. Rappazzo CG, Tse LV, Kaku CI, Wrapp D, Sakharkar M, Huang D, et al. Broad
and potent activity against SARS-like viruses by an engineered human
monoclonal antibody. Science. 2021;71(6531):8239. https://doi.org/10.1126/
science.abf4830.
91. Andreano E, Nicastri E, Paciello I, Pileri P, Manganaro N, Piccini G, et al.
Extremely potent human monoclonal antibodies from COVID-19
convalescent patients. Cell. 2021;184(7):18211835.e16. https://doi.org/10.1
016/j.cell.2021.02.035.
92. De Gasparo R, Pedotti M, Simonelli L, Nickl P, Muecksch F, Cassaniti I, et al.
Bispecific IgG neutralizes SARS-CoV-2 variants and prevents escape in mice.
Nature. 2021.
93. Brenner S, Jacob F, Meselson M. An unstable intermediate carrying
information from genes to ribosomes for protein synthesis. Nature. 1961;
190(4776):57681. https://doi.org/10.1038/190576a0.
94. Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, et al. Direct
gene transfer into mouse muscle in vivo. Science. 1990;247(4949 Pt 1):1465
8. https://doi.org/10.1126/science.1690918.
95. Malone RW, Felgner PL, Verma IM. Cationic liposome-mediated RNA
transfection. Proc Natl Acad Sci U S A. 1989;86(16):607781. https://doi.org/1
0.1073/pnas.86.16.6077.
96. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in
vaccinology. Nat Rev Drug Discov. 2018;17(4):26179. https://doi.org/10.103
8/nrd.2017.243.
97. Tourrière H, Chebli K, Tazi J. mRNA degradation machines in eukaryotic
cells. Biochimie. 2002;84(8):82137. https://doi.org/10.1016/s0300-9084(02
)01445-1.
98. Sahin U, Derhovanessian E, Miller M, Kloke BP, Simon P, Löwer M, et al.
Personalized RNA mutanome vaccines mobilize poly-specific therapeutic
immunity against cancer. Nature. 2017;547(7662):2226. https://doi.org/10.1
038/nature23003.
99. Linares-Fernández S, Lacroix C, Exposito JY, Verrier B. Tailoring mRNA
vaccine to balance innate/adaptive immune response. Trends Mol Med.
2020;26(3):31123. https://doi.org/10.1016/j.molmed.2019.10.002.
100. Martinon F, Krishnan S, Lenzen G, Magné R, Gomard E, Guillet JG, et al.
Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-
entrapped mRNA. Eur J Immunol. 1993;23(7):171922. https://doi.org/10.1
002/eji.1830230749.
101. Walsh EE, Frenck RW, Falsey AR, Kitchin N, Absalon J, Gurtman A, et al. Safety
and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J
Med. 2020;383(25):243950. https://doi.org/10.1056/NEJMoa2027906.
102. Walsh EE, Frenck R, Falsey AR, Kitchin N, Absalon J, Gurtman A, et al. RNA-
based COVID-19 vaccine BNT162b2 selected for a pivotal efficacy study.
medRxiv. 2020.
Novelli et al. Human Genomics (2021) 15:27 Page 12 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
103. Mulligan MJ, Lyke KE, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al.
Phase I/II study of COVID-19 RNA vaccine BNT162b1 in adults. Nature. 2020;
586(7830):58993. https://doi.org/10.1038/s41586-020-2639-4.
104. Banerji A, Wickner PG, Saff R, Stone CA Jr, Robinson LB, Long AA, et al. mRNA
vaccines to prevent COVID-19 disease and reported allergic reactions: current
evidence and approach. J Allergy Clin Immunol Pract. 2020.
105. Zhang NN, Li XF, Deng YQ, Zhao H, Huang YJ, Yang G, et al. A thermostable
mRNA vaccine against COVID-19. Cell. 2020;182(5):127183.e16. https://doi.
org/10.1016/j.cell.2020.07.024.
106. Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN,
et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J
Med. 2020;383(20):192031. https://doi.org/10.1056/NEJMoa2022483.
107. Corbett KS, Flynn B, Foulds KE, Francica JR, Boyoglu-Barnum S, Werner AP,
et al. Evaluation of the mRNA-1273 vaccine against SARS-CoV-2 in
nonhuman primates. N Engl J Med. 2020;383(16):154455. https://doi.org/1
0.1056/NEJMoa2024671.
108. Anderson EJ, Rouphael NG, Widge AT, Jackson LA, Roberts PC, Makhene M,
et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in
older adults. N Engl J Med. 2020;383(25):242738. https://doi.org/10.1056/
NEJMoa2028436.
109. Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, et al. Efficacy and
safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med. 2020.
110. Polack FP, Thomas SJ, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al.
Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med.
2020;383(27):260315. https://doi.org/10.1056/NEJMoa2034577.
111. Widge AT, Rouphael NG, Jackson LA, Anderson EJ, Roberts PC, Makhene M,
et al. Durability of responses after SARS-CoV-2 mRNA-1273 vaccination. N
Engl J Med. 2020;384(1):802. https://doi.org/10.1056/NEJMc2032195.
112. Thompson MG, Burgess JL, Naleway AL, Tyner HL, Yoon SK, Meece J, et al.
Interim estimates of vaccine effectiveness of BNT162b2 and mRNA-1273
COVID-19 vaccines in preventing SARS-CoV-2 infection among health care
personnel, first responders, and other essential and frontline workers eight
U.S. locations, December 2020 march 2021. MMWR Centers Dis Contr Prev
(CDC) Early Release. 2021;70.
113. Mascola JR, Fauci AS. Novel vaccine technologies for the 21st century. Nat
Rev Immunol. 2020;20(2):878. https://doi.org/10.1038/s41577-019-0243-3.
114. Blanchard EL, Vanover D, Bawage SS, Tiwari PM, Rotolo L, Beyersdorf J, et al.
Treatment of influenza and SARS-CoV-2 infections via mRNA-encoded
Cas13a in rodens. Nat Biotechnol. 2021.
115. Di Giorgio S, Martignano F, Torcia MG, Mattiuz G, Conticello SG. Evidence
for host-dependent RNA editing in the transcriptome of SARS-CoV-2. Sci
Adv. 2020;6(25):eabb5813. https://doi.org/10.1126/sciadv.abb5813.
116. Xie X, Liu Y, Liu J, Zhang X, Zou J, Fontes-Garfias CR, et al. Neutralization of
SARS-CoV-2 spike 69/70 deletion, E484K, and N501Y variants by BNT162b2
vaccine-elicited sera. bioRxiv. 2021.01.27.427998.
117. Wang Z, Schmidt F, Weisblum Y, Muecksch F, Barnes CO, Finkin S, et al.
mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants.
bioRxiv.2021.01.15.426911.
118. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S,
et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked
by a clinically proven protease inhibitor. Cell. 2020;181(2):271280.e8.
https://doi.org/10.1016/j.cell.2020.02.052.
119. Novelli G, Liu J, Biancolella M, Alonzi T, Novelli A, Patten JJ, et al. Inhibition
of HECT E3 ligases as potential therapy for COVID-19. Cell Death Dis. 2021;
12(4):310. https://doi.org/10.1038/s41419-021-03513-1.
PublishersNote
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Novelli et al. Human Genomics (2021) 15:27 Page 13 of 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The coronavirus disease 2019 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), rapidly became one of the most important and urgent health issues in the beginning of 2020 [1,2]. Since its emergence, it has continued to have a profound effect on global socioeconomic aspects and has affected the lives of hundreds of millions of people, worldwide. ...
... Genetic variation in TLR-encoding genes has been linked to COVID-19's significant respiratory symptoms [14,15]. Several studies have discovered different TLR genes related to COVID-19 susceptibility through genome-wide association studies or specific gene analysis [16,17]. The following polymorphisms are associated with the prognosis and susceptibility to COVID-19 infection: TLR3 rs3775290, TLR4 Asp299Gly, Thr399Ile, and TLR9 [16]. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a single-stranded RNA virus. Toll-like receptor 7 (TLR7) recognizes single-stranded RNA viruses. The TLR7 gene plays a critical role in the human innate and adaptive immune response to SARS-CoV-2 infections. Genetic factors probably affect SARS-CoV-2 infection susceptibility. In the current study, our aim was to search for genetic variations associated with COVID-19 patients in the TLR7 gene of a Korean population. We designed five gene-specific primers to cover the coding region of the human TLR7 gene. Using amplicon sequencing, we screened the genetic polymorphisms in the coding region of the TLR7 gene in COVID-19 patients and healthy controls. The genotype frequencies, allele frequencies, and Hardy–Weinberg equilibrium (HWE) were examined. We identified a low-frequency synonymous single nucleotide polymorphism (SNP) (rs864058) in the coding region of the TLR7 gene. There were no significant differences in the genotype or allele frequencies of the TLR7 rs864058 polymorphism between COVID-19 female patients and healthy controls (p = 1.0). In conclusion, TLR7 (rs864058) polymorphism is low frequency in Korean populations and is not associated with SARS-CoV-2 infection.
... The Omicron variant was identified in Botswana and is the most highly mutated strain, with twice as many mutations than the Delta variant [121,122]. The incidence of breakthrough cases during the Delta variant wave was high; however, despite the Delta variant being characterized as more aggressive in its symptomatology than prior strains [123,124], the disease outcome in vaccinated individuals was better than in those who were unvaccinated [125][126][127]. Intriguingly, during the Delta variant wave of COVID-19, viral loads in fully vaccinated individuals were as high as those seen in unvaccinated individuals, indicating that vaccination was reducing the risk of severe illness without reducing host cell viral infection and without blunting the production of viral particles in infected individuals [117,118,[128][129][130][131]. ...
Article
Full-text available
Anti-SARS-CoV-2 vaccines have played a pivotal role in reducing the risk of developing severe illness from COVID-19, thus helping end the COVID-19 global public health emergency after more than three years. Intriguingly, as SARS-CoV-2 variants emerged, individuals who were fully vaccinated did get infected in high numbers, and viral loads in vaccinated individuals were as high as those in the unvaccinated. However, even with high viral loads, vaccinated individuals were significantly less likely to develop severe illness; this begs the question as to whether the main effect of anti-SARS-CoV-2 vaccines is to confer protection against severe illness or immunity against infection. The answer to this question is consequential, not only to the understanding of how anti-SARS-CoV-2 vaccines work, but also to public health efforts against existing and novel pathogens. In this review, we argue that immune system sensitization-desensitization rather than sterilizing immunity may explain vaccine-mediated protection against severe COVID-19 illness even when the SARS-CoV-2 viral load is high. Through the lessons learned from COVID-19, we make the case that in the disease’s aftermath, public health agencies must revisit healthcare policies, including redefining the term “vaccine effectiveness.”
... Despite efforts, severe outcomes cannot be eluded in all cases, even in vaccinated subjects, as recently reported in literature [91]. The ease with which currently available mRNA vaccines [92] can be updated has made it possible to deal with the new variants of the virus in a timely manner [93,94]. ...
Article
Full-text available
Three and a half years after the pandemic outbreak, now that WHO has formally declared that the emergency is over, COVID-19 is still a significant global issue. Here, we focus on recent developments in genetic and genomic research on COVID-19, and we give an outlook on state-of-the-art therapeutical approaches, as the pandemic is gradually transitioning to an endemic situation. The sequencing and characterization of rare alleles in different populations has made it possible to identify numerous genes that affect either susceptibility to COVID-19 or the severity of the disease. These findings provide a beginning to new avenues and pan-ethnic therapeutic approaches, as well as to potential genetic screening protocols. The causative virus, SARS-CoV-2, is still in the spotlight, but novel threatening virus could appear anywhere at any time. Therefore, continued vigilance and further research is warranted. We also note emphatically that to prevent future pandemics and other world-wide health crises, it is imperative to capitalize on what we have learnt from COVID-19: specifically, regarding its origins, the world’s response, and insufficient preparedness. This requires unprecedented international collaboration and timely data sharing for the coordination of effective response and the rapid implementation of containment measures.
... The results showed that the vaccinated group exhibited a significant reduction in certain clinical symptoms, such as fever and cough. Also, they found that vaccination could reduce the severity of disease (consistent with the study by Giuseppe et al. [55]) also found and more patients with asymptomatic infection in the vaccinated group. ...
Article
Background: The coronavirus (COVID-19) pandemic caused enormous adverse socioeconomic impacts worldwide. Evidence suggests that the diagnostic accuracy of clinical features of COVID-19 may vary among different populations. Methods: We conducted a systematic review and meta-analysis of studies from PubMed, Embase, Cochrane Library, Google Scholar, and the WHO Global Health Library for studies evaluating the accuracy of clinical features to predict and prognosticate COVID-19. We used the National Institutes of Health Quality Assessment Tool to evaluate the risk of bias, and the random-effects approach to obtain pooled prevalence, sensitivity, specificity, and likelihood ratios. Results: Among the 189 included studies (53 659 patients), fever, cough, diarrhoea, dyspnoea, and fatigue were the most reported predictors. In the later stage of the pandemic, the sensitivity in predicting COVID-19 of fever and cough decreased, while the sensitivity of other symptoms, including sputum production, sore throat, myalgia, fatigue, dyspnoea, headache, and diarrhoea, increased. A combination of fever, cough, fatigue, hypertension, and diabetes mellitus increases the odds of having a COVID-19 diagnosis in patients with a positive test (positive likelihood ratio (PLR) = 3.06)) and decreases the odds in those with a negative test (negative likelihood ratio (NLR) = 0.59)). A combination of fever, cough, sputum production, myalgia, fatigue, and dyspnea had a PLR = 10.44 and an NLR = 0.16 in predicting severe COVID-19. Further updating the umbrella review (1092 studies, including 3 342 969 patients) revealed the different prevalence of symptoms in different stages of the pandemic. Conclusions: Understanding the possible different distributions of predictors is essential for screening for potential COVID-19 infection and severe outcomes. Understanding that the prevalence of symptoms may change with time is important to developing a prediction model.
... The COVID-19 pandemic emergency upended life and particularly education worldwide. The sudden pivot to online delivery brought many challenges but also revealed technological opportunities that are advantageous and will remain intact even after the return to in-person education [44][45][46]. ...
Article
Full-text available
A course-based undergraduate research experience (CURE) is described, both in its original, field-based and in-person design, as well as its adaptation to online delivery during the COVID-19 pandemic. The CURE experience was centered around the topic of microbes attaching to ocean plastic debris, and included connecting nontraditional undergraduate students attending a primarily teaching institution to personnel of a research-intensive institution. The CURE was implemented in nonmajors and majors’ general biology laboratory courses, as well as in an introductory microbiology lab course for health majors. Student perception of the CURE was assessed quantitatively through self-administered retrospective surveys, and qualitatively using open-ended survey questions and focus group interviews. Survey questions were grouped into four themes: (1) research at the research institution, (2) research at the teaching institution, (3) laboratory skills, and (4) interest in science. To assess impact of the CURE by modality, major and course level, theme scores were analyzed using ANOVAs (α = 0.05). Changes in student perception of learning were measured by comparing the “before” and “after” survey scores in each theme. The main source of variation in the model was time, followed by theme and major, while modality had a negligible effect. Overall, there were increases in student perception of learning across all themes across modality, major, and course level; however, not all were statistically significant. Highest gains were observed among nonmajors, while those of the majors’ upper division courses were the lowest. On the other hand, majors’ overall scores were the highest. Health majors’ survey gains were mostly between those of nonmajors and majors. Qualitative data contextualized the quantitative results. The online adaptation was a valuable alternative, especially for nonmajors, as it expanded the range of students reached, with results similar to the in-person alternative. Preliminary data show a positive effect of the CURE in student success majoring in biology.
... Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) was declared a pandemic by the World Health Organization (WHO) on 11 March 2020. SARS-CoV2 vaccines were rapidly developed following the global emergence of this pandemic, and the vaccine benefit has been shown to significantly lower mortality rates [1][2][3]. However, the emergence of SARS-CoV2 variants of concern (VOC) reduced potency of existing prototype vaccines are driving the development of vaccines that could provide protection against VOC [4][5][6][7][8]. ...
Article
After severe acute respiratory syndrome coronavirus-2 (SARS-CoV2) made the world tremble with a global pandemic, SARS-CoV2 vaccines were developed. However, due to the coronavirus's intrinsic nature, new variants emerged, such as Delta and Omicron, refractory to the vaccines derived using the original Wuhan strain. We developed an HERV-enveloped recombinant baculoviral DNA vaccine against SARS-CoV2 (AcHERV-COVID19S). A non-replicating recombinant baculovirus that delivers the SARS-CoV2 spike gene showed a protective effect against the homologous challenge in a K18-hACE2 Tg mice model; however, it offered only a 50 % survival rate against the SARS-CoV2 Delta variant. Therefore, we further developed the AcHERV-COVID19 Delta vaccine (AcHERV-COVID19D). The AcHERV-COVID19D induced higher neutralizing antibodies against the Delta variant than the prototype or Omicron variant. On the other hand, cellular immunity was similarly high for all three SARS-CoV2 viruses. Cross-protection experiments revealed that mice vaccinated with the AcHERV-COVID19D showed 100 % survival upon challenge with Delta and Omicron variants and 71.4 % survival against prototype SARS-CoV2. These results support the potential of the viral vector vaccine, AcHERV-COVID19D, in preventing the spread of coronavirus variants such as Omicron and SARS-CoV2 variants.
... We found that vaccination could reduce the severity of disease, which was consistent with the study of Giuseppe, 72 and more patients with asymptomatic infection were found in the vaccinated group in our study. What plays a central role in vaccines are the antigens. ...
Article
Full-text available
Objective: With the global epidemic of coronavirus disease 2019 (COVID-19), vaccination rates are increasing globally. This study evaluated the relevant clinical manifestations of vaccinated COVID-19 patients. Methods: We searched carefully in 11 databases such as PubMed, Embase, Scopus, Cochrane Library, Web of Science, Ovid, China National Knowledge Infrastructure Database, Wan Fang Data, Sinomed, VIP Database, and Reading Showing Database up to 26 March 2022. To search for articles that have described the characteristics of vaccinated patients including epidemiological and clinical symptoms. Statistical analysis of the extracted data using STATA 14.0. Results: A total of 58 articles and 263,708 laboratory-confirmed COVID-19 patients were included. Most of the patients in the vaccinated group had more asymptomatic infection and fewer severe illnesses. There were significant differences in ethnicity, and strain infected with COVID-19, and comorbidities (hyperlipidemia, diabetes, obesity, kidney disease, immunocompromised, cardiovascular disease, and tumor) and symptoms (fever, cough, gastrointestinal symptoms, neurological symptoms, and dysgeusia/anosmia) between vaccinated group and unvaccinated group. Oxygen support, use of steroid, days in hospital, hospital treatment, ICU treatment, death, and poor prognosis were also significantly different. Conclusion: Compared with the vaccinated group, patients in the unvaccinated group had a more severe clinical manifestations. Vaccines are also protective for infected people.
... An interdisciplinary and multimodal approach is needed to meet the challenges posed by the COVID-19 pandemic. It is already well known that in various manifestations of infectious diseases, in addition to factors such as the general state of health, age, sex, and virulence of the pathogen, the individual genetic constellation plays a key role [13,14]. ...
Article
Background Exploring the pathogenetic mechanisms behind severe lung damage in COVID-19 is crucial. In this study, we decided to focus on two molecular markers that affect surfactant metabolism and lung development: the surfactant protein B (SFTPB) and the glucocorticoid receptor (NR3C1) genes. The aim of our study was to determine the effect of SFTPB (rs11130866) and NR3C1 (rs41423247) gene variants on the course of the disease in patients with COVID-19, and the treatment measures they required. Methods The study group included 58 patients with a diagnosis of severe “viral COVID-19 pneumonia.” Determination of SFTPB and NR3C1 gene variants was performed using the PCR-RFLP method. Results Our results indicate that the presence of the SFTPB gene CC genotype increases the risk of developing acute respiratory distress syndrome in patients with COVID-19 (χ2 = 4.03, p = 0.045, OR = 3.90 [1.19–12.78]). However, patients with the SFTPB gene TT genotype required respiratory support for a shorter period of time. Patients with the NR3C1 gene CC genotype underwent a longer glucocorticoid therapy. Moreover, for patients with the CC genotype, a longer stay in the intensive care unit was detected before lethal outcome. Conclusions The obtained results confirm the influence of the SFTPB (rs11130866) and NR3C1 (rs41423247) gene variants on the therapy, course, and severity of the disease in patients with COVID-19. Of course, these results require further study, analysis, and larger, complex, systematic research.
Preprint
Full-text available
Genetic information is encoded in a linear sequence of nucleotides, represented by letters ranging from thousands to billions. Mutations refer to changes in the DNA or RNA nucleotide sequence. Thus, mutation detection is vital in all areas of biology and medicine. Careful monitoring of virulence-enhancing mutations is essential. However, an enormous amount of classical computing power is required to analyze genetic sequences of this size. Inspired by human perception of vision and pixel representation of images on quantum computers, we leverage these techniques to implement a pairwise sequence analysis. The methodology has a potential advantage over classical approaches and can be further applied to identify mutations and other modifications in genetic sequences. We present a method to display and analyze the similarity between two genome sequences on a quantum computer where a similarity score is calculated to determine the similarity between nucleotides.
Article
Full-text available
The emergence and spread of SARS-CoV-2 lineage B.1.1.7, first detected in the United Kingdom, has become a global public health concern because of its increased transmissibility. Over 2500 COVID-19 cases associated with this variant have been detected in the US since December 2020, but the extent of establishment is relatively unknown. Using travel, genomic, and diagnostic data, we highlight that the primary ports of entry for B.1.1.7 in the US were in New York, California, and Florida. Furthermore, we found evidence for many independent B.1.1.7 establishments starting in early December 2020, followed by interstate spread by the end of the month. Finally, we project that B.1.1.7 will be the dominant lineage in many states by mid to late March. Thus, genomic surveillance for B.1.1.7 and other variants urgently needs to be enhanced to better inform the public health response.
Article
Full-text available
Messenger RNA (mRNA) BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna) COVID-19 vaccines have been shown to be effective in preventing symptomatic COVID-19 in randomized placebo-controlled Phase III trials (1,2); however, the benefits of these vaccines for preventing asymptomatic and symptomatic SARS-CoV-2 (the virus that causes COVID-19) infection, particularly when administered in real-world conditions, is less well understood. Using prospective cohorts of health care personnel, first responders, and other essential and frontline workers* in eight U.S. locations during December 14, 2020-March 13, 2021, CDC routinely tested for SARS-CoV-2 infections every week regardless of symptom status and at the onset of symptoms consistent with COVID-19-associated illness. Among 3,950 participants with no previous laboratory documentation of SARS-CoV-2 infection, 2,479 (62.8%) received both recommended mRNA doses and 477 (12.1%) received only one dose of mRNA vaccine.† Among unvaccinated participants, 1.38 SARS-CoV-2 infections were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) per 1,000 person-days.§ In contrast, among fully immunized (≥14 days after second dose) persons, 0.04 infections per 1,000 person-days were reported, and among partially immunized (≥14 days after first dose and before second dose) persons, 0.19 infections per 1,000 person-days were reported. Estimated mRNA vaccine effectiveness for prevention of infection, adjusted for study site, was 90% for full immunization and 80% for partial immunization. These findings indicate that authorized mRNA COVID-19 vaccines are effective for preventing SARS-CoV-2 infection, regardless of symptom status, among working-age adults in real-world conditions. COVID-19 vaccination is recommended for all eligible persons.
Article
Full-text available
Neutralizing antibodies targeting the receptor binding domain (RBD) of the SARS-CoV-2 Spike (S) are among the most promising approaches against coronavirus disease 2019 (COVID-19)1,2. We developed a bispecific, IgG1-like molecule (CoV-X2) based on two antibodies derived from COVID-19 convalescent donors, C121 and C1353. CoV-X2 simultaneously binds two independent sites on the RBD and, unlike its parental antibodies, prevents detectable S binding to Angiotensin-Converting Enzyme 2 (ACE2), the virus cellular receptor. Furthermore, CoV-X2 neutralizes SARS-CoV-2 and its variants of concern, as well as the escape mutants generated by the parental monoclonals. In a novel animal model of SARS-CoV-2 infection with lung inflammation, CoV-X2 protects mice from disease and suppresses viral escape. Thus, simultaneous targeting of non-overlapping RBD epitopes by IgG-like bispecific antibodies is feasible and effective, combining into a single molecule the advantages of antibody cocktails.
Article
Full-text available
SARS-CoV-2 is responsible for the ongoing world-wide pandemic which has already taken more than two million lives. Effective treatments are urgently needed. The enzymatic activity of the HECT-E3 ligase family members has been implicated in the cell egression phase of deadly RNA viruses such as Ebola through direct interaction of its VP40 Protein. Here we report that HECT-E3 ligase family members such as NEDD4 and WWP1 interact with and ubiquitylate the SARS-CoV-2 Spike protein. Furthermore, we find that HECT family members are overexpressed in primary samples derived from COVID-19 infected patients and COVID-19 mouse models. Importantly, rare germline activating variants in the NEDD4 and WWP1 genes are associated with severe COVID-19 cases. Critically, I3C, a natural NEDD4 and WWP1 inhibitor from Brassicaceae , displays potent antiviral effects and inhibits viral egression. In conclusion, we identify the HECT family members of E3 ligases as likely novel biomarkers for COVID-19, as well as new potential targets of therapeutic strategy easily testable in clinical trials in view of the established well-tolerated nature of the Brassicaceae natural compounds.
Article
Full-text available
It is plausible that variants in the ACE2 and TMPRSS2 genes might contribute to variation in COVID-19 severity and that these could explain why some people become very unwell whereas most do not. Exome sequence data was obtained for 49,953 UK Biobank subjects, of whom 82 had tested positive for SARS-CoV-2 and could be presumed to have severe disease. A weighted burden analysis was carried out using SCOREASSOC to determine whether there were differences between these cases and the other sequenced subjects in the overall burden of rare, damaging variants in ACE2 or TMPRSS2. There were no statistically significant differences in weighted burden scores between cases and controls for either gene. There were no individual DNA sequence variants with a markedly different frequency between cases and controls. Whether there are small effects on severity, or whether there might be rare variants with major effect sizes, would require studies in much larger samples. Genetic variants affecting the structure and function of the ACE2 and TMPRSS2 proteins are not the main explanation for why some people develop severe symptoms in response to infection with SARS-CoV-2. This research was conducted using the UK Biobank Resource.
Article
Full-text available
Objective To establish whether there is any change in mortality from infection with a new variant of SARS-CoV-2, designated a variant of concern (VOC-202012/1) in December 2020, compared with circulating SARS-CoV-2 variants. Design Matched cohort study. Setting Community based (pillar 2) covid-19 testing centres in the UK using the TaqPath assay (a proxy measure of VOC-202012/1 infection). Participants 54 906 matched pairs of participants who tested positive for SARS-CoV-2 in pillar 2 between 1 October 2020 and 29 January 2021, followed-up until 12 February 2021. Participants were matched on age, sex, ethnicity, index of multiple deprivation, lower tier local authority region, and sample date of positive specimens, and differed only by detectability of the spike protein gene using the TaqPath assay. Main outcome measure Death within 28 days of the first positive SARS-CoV-2 test result. Results The mortality hazard ratio associated with infection with VOC-202012/1 compared with infection with previously circulating variants was 1.64 (95% confidence interval 1.32 to 2.04) in patients who tested positive for covid-19 in the community. In this comparatively low risk group, this represents an increase in deaths from 2.5 to 4.1 per 1000 detected cases. Conclusions The probability that the risk of mortality is increased by infection with VOC-202012/01 is high. If this finding is generalisable to other populations, infection with VOC-202012/1 has the potential to cause substantial additional mortality compared with previously circulating variants. Healthcare capacity planning and national and international control policies are all impacted by this finding, with increased mortality lending weight to the argument that further coordinated and stringent measures are justified to reduce deaths from SARS-CoV-2.
Preprint
Full-text available
We identified a novel SARS-CoV-2 variant by viral whole-genome sequencing of 2,172 remnant nasal/nasopharyngeal swab samples from 44 counties in California. Named B.1.427/B.1.429 or 20C/L452R, the variant emerged around May 2020 and increased from 0% to >50% of sequenced cases from September 1, 2020 to January 29, 2021, exhibiting an estimated 18.6-24% increase in transmissibility relative to wild-type circulating strains. This variant is characterized by three mutations in the spike protein, including a L452R substitution in the receptor-binding domain. Our analyses revealed 2-fold increased B.1.427/B.1.429 viral shedding in vivo and increased L452R pseudovirus infection of cell cultures and lung organoids, albeit decreased relative to pseudoviruses carrying the N501Y mutation found in SARS-CoV-2 variants of concern (B.1.1.7, B.1.351, and P.1 lineages). Antibody neutralization assays showed 4.0 to 6.7-fold and 2.0-fold decreases in neutralizing titers from convalescent patients and vaccine recipients, respectively. The increased prevalence of a more transmissible variant in California associated with decreased antibody neutralization warrants further investigation.
Article
Neutralizing antibodies (nAbs) hold promise as therapeutics against COVID-19. Here, we describe protein engineering and modular design principles that have led to the development of synthetic bivalent and tetravalent nAbs against SARS-CoV-2. The best nAb targets the host receptor binding site of the viral S-protein and tetravalent versions block entry with a potency exceeding bivalent nAbs by an order of magnitude. Structural studies show that both the bivalent and tetravalent nAbs can make multivalent interactions with a single S-protein trimer, consistent with the avidity and potency of these molecules. Significantly, we show that the tetravalent nAbs show increased tolerance to potential virus escape mutants and an emerging variant of concern. Bivalent and tetravalent nAbs can be produced at large-scale and are as stable and specific as approved antibody drugs. Our results provide a general framework for enhancing antiviral therapies against COVID-19 and related viral threats, and our strategy can be applied to virtually any antibody drug.