ArticlePDF AvailableLiterature Review

Abstract and Figures

COVID-19, which is caused by the SARS-CoV-2, has ravaged the world for the past 2 years. Here, we review the current state of research into the disease with focus on its history, human genetics and genomics and the transition from the pandemic to the endemic phase. We are particularly concerned by the lack of solid information from the initial phases of the pandemic that highlighted the necessity for better preparation to face similar future threats. On the other hand, we are gratified by the progress into human genetic susceptibility investigations and we believe now is the time to explore the transition from the pandemic to the endemic phase. The latter will require worldwide vigilance and cooperation, especially in emerging countries. In the transition to the endemic phase, vaccination rates have lagged and developed countries should assist, as warranted, in bolstering vaccination rates worldwide. We also discuss the current status of vaccines and the outlook for COVID-19.
This content is subject to copyright. Terms and conditions apply.
Biancolellaetal. Human Genomics (2022) 16:19
https://doi.org/10.1186/s40246-022-00392-1
REVIEW
COVID-19 2022 update: transition
ofthepandemic totheendemic phase
Michela Biancolella1†, Vito Luigi Colona2†, Ruty Mehrian‑Shai3, Jessica Lee Watt4, Lucio Luzzatto5,6,
Giuseppe Novelli2,7,8,10*† and Juergen K. V. Reichardt9†
Abstract
COVID‑19, which is caused by the SARS‑CoV‑2, has ravaged the world for the past 2 years. Here, we review the current
state of research into the disease with focus on its history, human genetics and genomics and the transition from the
pandemic to the endemic phase. We are particularly concerned by the lack of solid information from the initial phases
of the pandemic that highlighted the necessity for better preparation to face similar future threats. On the other hand,
we are gratified by the progress into human genetic susceptibility investigations and we believe now is the time
to explore the transition from the pandemic to the endemic phase. The latter will require worldwide vigilance and
cooperation, especially in emerging countries. In the transition to the endemic phase, vaccination rates have lagged
and developed countries should assist, as warranted, in bolstering vaccination rates worldwide. We also discuss the
current status of vaccines and the outlook for COVID‑19.
© The Author(s) 2022. 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:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Introduction
By the end of December 2019, information began to cir-
culate on an alarming form of pneumonia, of unknown
etiology, that was afflicting the district of Wuhan, China.
Two years later, it is of common knowledge that was
the beginning of a pandemic, as declared by the World
Health Organization (WHO) [1], triggered by the new
Severe Acute Respiratory Syndrome Coronavirus-2
(SARS-CoV-2), the causative agent of Coronavirus Dis-
ease 2019 (COVID-19).
While facing the critical times of the manifestation
of a "fourth wave,” amenable to the appearance of new
variants [24], there has been exponential growth of
new data. ese data explore the genetics of the virus,
the interaction with the host, as well as short-term and
long-term clinical manifestations. Due to this, we believe
there is need to provide an updated overview to uphold
the commitment made in our latest Editorial [5].
In recent months, we globally experienced a rise in
daily cases, contributing to a total of 527,971,809 cases
and 6,284,871 deaths since the beginning of the pan-
demic (Johns Hopkins University, CSSE, accessed on
2022) [6]. Circulating variants have been supplanted
by the new variant of concern (VOC) SARS-CoV-2
B.1.1.529 (Omicron) and its sub-variants [7, track-
ing website accessed on April 19, 2022], as SARS-
CoV-2 BA.1 and BA.2, which are now dominant in
the USA (Centers for Disease Control and Prevention.
COVID Data Tracker. Atlanta, GA: US Department of
Health and Human Services, CDC; https:// covid. cdc.
gov/ covid- data- track er, accessed on April 19, 2022)
[8] and globally. Characterized by a greater ability to
evade immune responses acquired through infection
with a different strain [9] or through vaccination [10],
this variant seems to be able to change the profile of
current outbreak and, unlike in the previous waves, a
higher rate of reinfections is reported [11, 12]. ese
early data, still under investigation, urge the scientific
Open Access
Michela Biancolella, Vito Luigi Colona, Giuseppe Novelli and Juergen K.V.
Reichardt contributed equally.
*Correspondence: novelli@med.uniroma2.it
10 Department of Biomedicine and Prevention, School of Medicine
and Surgery, Via Montpellier 1, 00133 Rome, Italy
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 12
Biancolellaetal. Human Genomics (2022) 16:19
community toward the search for therapeutic and
preventive tools that can counter this evolutionary
mechanism.
In our review “COVID-19 one year into the pan-
demic: from genetics and genomics to therapy, vac-
cination, and policy” [13], we strongly claimed the
determining role of vaccines as the most valuable aid
to halt the spreading of SARS-CoV-2. We can now
affirm that the increasing vaccination rate, with a total
of 11.184.961.194 doses administered (Johns Hopkins
University, CSSE, accessed on April 19, 2022) [6], is
contributing to the containment of hospitalizations
and deaths in the population affected by COVID-19
[1416].
e importance of a homogeneous and universal dis-
tribution of vaccines is becoming more evident and
incisive in hindering the appearance of new variants.
In this regard, the disparities between advanced and
developing countries seem to worsen, and this results
in the inability to cope with new manifestations, as
highlighted by the appearance of the SARS-CoV-2
B.1.1.529 variant [17].
Origin andcurrent state
COVID-19 was first reported in 2019 [18, 19]. It has now
raged worldwide for more than 2years, affecting every
corner of our globe with no clear indication how the pan-
demic started. An intelligible understanding of the ori-
gins of this pandemic is critical to be better prepared in
the future. We lament that a panel proposed by the WHO
in 2021 to achieve this goal has not yet reached signifi-
cant conclusions [20].
While the pandemic is still raging, sprouts of hope
have emerged that we may be transitioning into the
endemic phase [21]. e pervasive Omicron variant, cur-
rently predominant, may lead to this course [22]. How-
ever, overly exuberant enthusiasm must be tempered by
a sense of reality and concern for emerging countries
[21, 23, 24]. Developed countries must remain vigilant
and assist emerging countries in the fight against SARS-
CoV-2, with the aim of detecting new variants of concern
Fig. 1 A comical view of the history of COVID‑19. A few translations: “vorhersehbar” = predictable and “war ja klar” = obviously or sure or of course.
Reproduced with permission (Mira Nagel)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 12
Biancolellaetal. Human Genomics (2022) 16:19
(Fig.1), investigating animal reservoirs and sharing diag-
nostic tools, surveillance and therapies [25].
Clinical manifestation ofCOVID‑19 andpost‑acute
(long) COVID‑19
It is now known that COVID-19 is a multisystem condi-
tion that largely involves the respiratory system. It starts
as an upper respiratory tract infection that subsequently
affects the lungs and establishes, in the most severe cases,
interstitial pneumonia (showing the diagnostic ground
glass appearance, through CT investigation), severe res-
piratory failure, systemic inflammatory response and
multi-organ dysfunction. Classic symptoms of the dis-
ease are listed as fever, asthenia, dry cough, nasal conges-
tion and breathing difficulties.
Signs and symptoms, however, can affect several
organs. Other systems can be involved, such as the cen-
tral nervous system (hypo-anosmia, loss of the sense
of taste, speech disturbances, dizziness, alterations of
the consciousness and behavior, impaired walking and
maintenance of upright position, impaired hearing and
vision), the cardiovascular system (alterations in hemo-
stasis, arrhythmia, heart failure), the gastrointestinal sys-
tem (nausea, emesis, diarrhea, abdominal pain), the renal
system, neuromuscular (myalgia) and skin adnexa [26].
Clinical manifestation of infection is therefore
extremely heterogeneous, ranging from completely
asymptomatic or paucisymptomatic subjects to criti-
cally ill patients who require hospitalization and venti-
latory support in intensive care unit [27, 28]. Since the
beginning of the pandemic, the medical community has
been aware of the greater susceptibility of patients with
advanced age and comorbidities to the most serious
forms of the disease, but we now know that patients with
a younger age can also be critically affected [29].
Although virus-host interactions have been deeply
investigated [3032], the mechanisms underpinning a
longer persistence of the symptoms in some patients or
their recurrence (4 to 5weeks, or even 1year) after the
resolution of the disease remain to be understood [33].
e persistence of fatigue, headache, and anosmia, the
onset of anxiety and a depressive state are symptoms that
have recently been included in the so-called post-acute
(long) COVID-19 [26, 33, 34].
Like COVID-19, post-acute (long) COVID-19 is con-
figured as a systemic disease and therefore symptoms
are extremely varied and of difficult clinical interpreta-
tion. ey can occur singly or in combination, they can
be transient, intermittent or constant, and they can even
change over the course of the condition.
e systems involved in post-acute (long) COVID-19
are mainly respiratory, musculoskeletal, cardiovascular
and neurological [35].
Given the predominantly respiratory nature of the con-
dition, lungs are the organs susceptible to the most severe
outcomes, not only on a structural level (e.g., secondary
interstitial fibrosis, pulmonary hypertension) [36, 37], but
also on a functional level (e.g., reduced ventilatory capac-
ity, dyspnea, fatigue) [3739].
Respiratory sequelae have inevitably been shown to
have repercussions at neuromuscular levels. In fact, dys-
functions of both respiratory and skeletal muscles have
been described in about 40% of patients admitted to
intensive care units, resulting in persistent symptoms of
fatigue, weakness and shortness of breath [4042]. Fur-
thermore, it has been hypothesized that a direct muscle
affection of SARS-CoV-2 may be responsible for struc-
tural alterations, even in patients who have had an appar-
ently mild disease outcome [43].
e heart has been shown to be a target organ of the
systemic inflammatory response and subject to direct
damage from SARS-CoV-2 [35]. Specifically, the most
described cardiovascular complications refer to heart
failure, arrhythmias, peri-myocarditis, venous and arte-
rial thromboembolism and “reverse Tako-Tsubo” cardio-
myopathy [35, 44, 45].
Approximately 25% of patients who developed
COVID-19 experienced neurological disorders of vari-
ous degrees in the months following diagnosis [46]. e
most common and mild symptoms include headache
[47], disturbances in perception of taste and smell [48
50], “brain fog” and memory disorders [51]. Among the
major complications, however, those mostly described
were the presence of diffuse brain damage of inflam-
matory [52] or acute metabolic origins (toxic-metabolic
encephalopathies) [53], Guillain-Barré syndrome [54,
55], Miller-Fisher syndrome [56], ischemic vasculitis [57],
dysautonomic dysfunctions [55, 58, 59] and seizures. In
addition to these complications. we urge the scientific
community to deeply investigate mood disorders that
might develop on a psychological substrate in response to
stressors established during the pandemic period [46, 52,
60, 61]. Various mechanisms may underlie the neurologi-
cal implications of SARS-CoV-2 [35, 62]. e systemic
inflammatory response triggered in COVID-19 patients
could potentially accelerate the evolution of neurodegen-
erative processes by exacerbating chronic conditions pre-
sent at the time of infection but not yet manifest [35, 63,
64]. For example, the inflammatory response could exac-
erbate a refractory epileptic condition [65]. Several stud-
ies also reported direct damage of the brain tissue caused
by SARS-CoV-2 infection [35, 66, 67].
In a novel, quantitative, longitudinal imaging study
from Douaud et al. [68], authors analyzed brain scans
of 401 SARS-CoV-2 positive cases, acquired at two time
points (before and after testing positive for infection),
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 12
Biancolellaetal. Human Genomics (2022) 16:19
and 384 matching controls. Following the description of
hundreds of derived phenotypes, the comparison pro-
vided suggestions as to what could be further investigated
as effect caused by, or attributable to, the SARS-CoV-2
infection. A clear involvement of the olfactory cortex has
been detected through variations in tissue damage mark-
ers. e evaluation of gray matter decrease showed FDG
(fluorodeoxyglucose) hypometabolism in the orbitofron-
tal cortex, insula, parahippocampal gyrus, and anterior
cingulate cortex, suggesting a significant implication of
the regions connected to the piriform cortex. In conclu-
sion, the effect of reduction in the gray matter appears to
be generalized, with a greater relevance at the level of the
olfactory system.
However, to date, knowledge of the molecular mecha-
nisms and neurological consequences in the post-acute
(long) COVID-19 is still limited [69, 70].
In a broad sense, it has been hypothesized that post-
acute (long) COVID-19 can be considered a condition
characterized by a chronic persistence of low levels of
inflammatory cytokines [71]. According to this asser-
tion, it is likely that the activation of some cellular tran-
scription factors, including the nuclear factor erythroid
2 (NFE2)-related factor 2 (Nrf2), a possible therapeutic
target in several chronic neurodegenerative conditions
[7274], may have a role in increasing the expression of
enzymes capable of synthesizing glutathione, therefore
reducing the state of oxidative stress [71, 74, 75]. How-
ever, further data and trials are needed [76].
A recent study highlighted a significant formation
of blood micro-clots, both in the acute phase and in
the post-illness phase. ese micro-clots seem to show
resistance to the body’s fibrinolytic processes in patients
suffering from post-acute (long) COVID-19. Preliminary
results demonstrated the efficacy in reducing the symp-
toms of long COVID-19 patients through the administra-
tion of antiplatelet or anticoagulant therapy [77].
Understanding the signs and symptoms of the disease
and of post-acute (long) COVID-19 represents a major
current therapeutic challenge. is will allow us, in the
near future, not only to better elucidate the molecu-
lar mechanisms of our body’s response to SARS-CoV-2
infection, but also to identify therapeutic targets for an
increasingly personalized medicine.
Genetic susceptibility inthehost
Viruses, like other pathogens, are necessary, but not suffi-
cient, to trigger disease [78]. It therefore appears evident
that the individual host genome plays a fundamental role,
not only in the susceptibility to disease induced by the
infectious agent, but also in the individual response in
terms of severity of the phenotype or resistance to infec-
tion [78]. Numerous host genes have been identified in
the last two years that are active in susceptibility/resist-
ance to infectious diseases [5, 13, 7981]. Identifying
and qualifying these genes as prognostic and predictive
biomarkers is crucial to optimize patient management
and promote sustainable and rational public health (PH)
interventions. In addition, they contribute to clarify the
mechanisms and variability of the SARS-CoV-2 host–
pathogen interactions [81].
Common and rare variants have been identified in
different studies using a) classical Genome-Wide Asso-
ciation Studies (GWAS) and b) deep sequencing of
genes coding for protein referable to precise biochemi-
cal pathways involved in the pathogenesis of the infec-
tion. ese studies have made it possible to identify
alleles of increased susceptibility and/or partial resist-
ance to the COVID-19, in coding and non-coding
regions of genes. For example, a functional analysis of a
SNP (rs11385942), identified by GWAS on chromosome
3p21.31, demonstrated the involvement of the LZTFL1
protein (leucine zipper transcription factor like 1) [82],
which regulates ciliary localization in the BBSome com-
plex. is gene is mutated in Bardet-Biedl Syndrome
(BBS) (MIM#209,900), a ciliopathy characterized in
part by polydactyly, obesity, cognitive impairment,
hypogonadism, and kidney failure. LZTFL1 is highly
expressed in ciliated cells, including airway ciliated cells.
Its reduced expression leads to fewer airway ciliated
cells with shorter cilia, which could result in inefficient
viral airway clearance in COVID-19 patients. Similarly,
the SNP rs74956615, which maps on the chromosome
19p13.2 in the untranslated 3’ of the RAVER1 gene [82],
has been found to modulate the expression of RAV ER1
itself. is gene encodes for a ribonucleoprotein which
cooperates with cytoskeletal proteins vinculin/metavin-
culin and alpha-actinin to modulate alternative splicing
events. However, RAVER1 is a co-activator of MDA5
(IFIH1), which recognizes nucleic acids associated with
viral infections such as dsRNAs, including SARS-CoV-2,
and activates antiviral response genes, including IFNB1,
ICAM1, TNF and CCL5. A large human genetic study,
involving more than 49,000 COVID-19-affected individ-
uals and 2 million control subjects, identified 13 loci in
the human genome that affect COVID-19 susceptibility
and severity including 6 loci previously not reported [83].
In the regions mapped by this extensive GWAS, authors
identified more than 40 candidate genes, several of which
are involved in immune function or have known func-
tions in the lungs, suggesting that these may have impor-
tant effects on COVID-19. A suggestive susceptibility
locus on chromosome 12q22, has been recently detected
in ai population [84]. Genes mapped in this area
include EEA1 and LOC643339. EEA1 is involved in viral
entry into cells, while LOC643339 is a long non-coding
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 12
Biancolellaetal. Human Genomics (2022) 16:19
RNA. Intriguingly, EEA1 is involved in the entry of Afri-
can Swine Fever Virus via endosomal pathway [85]. Uti-
lizing a Phenome-Wide Association Study (PheWAS)
approach, Regan and Colleagues [86] identified novel
phenotypic associations with genes encoding proteins
active in the antiviral response and inflammatory pro-
cesses. ese genomic biomarkers can have pleiotropic
effects in COVID-19-related comorbidities (cardiovascu-
lar disease, autoimmune disease, arthropathy and endo-
crinopathy), which in turn increase the risk of severe
COVID-19.
Finally, a recent GWAS meta-analysis that considered
125,584 cases and over 2.5 million controls evaluating 60
studies from 25 countries included 11 new significant loci
at the genome level, in addition to those previously iden-
tified. Genes in the new loci include SFTPD, MUC5B and
ACE2, revealing convincing information on the suscepti-
bility and severity of the disease [83].
Candidate-gene approach, on the other hand, made it
possible to confirm and integrate the role of some specific
pathways and proteins in the pathogenesis of the disease.
Among the first candidate genes studied in SARS-CoV-2
infection were those coding for the HLA system, which
plays a crucial role in the immune response [87, 88].
Several studies have highlighted risk alleles capable of
influencing the clinical course of patients infected with
various RNA viruses (e.g., H1N1 influenza virus [89],
Hantaan virus [90] and SARS-CoV-1 [87]). Several stud-
ies have highlighted HLA alleles of susceptibility to
SARS-CoV-2 [28, 91]. However, these studies revealed
discrepancies due to different stratifications of patients
and controls and to the different frequency distribution
of the HLA alleles in the populations analyzed. Recently, a
large and accurate study described a potential association
of HLA-C*04:01 with severe clinical course of COVID-
19. Carriers of HLA-C*04:01 had twice the risk of need-
ing intubation when infected with SARS-CoV-2 [92].
Numerous other candidate genes have been analyzed on
the basis of their biological function during infection,
such as ACE2, TMPRSS2, DPP4, andFurin,involved in
the entry of the virus into cells, or genes active in the viral
egress such as WWP1 and NEDD4 [9397]. Curiously, an
association of VDR gene polymorphisms with COVID-
19 outcomes has been also detected [98]. e possi-
ble involvement of this receptor is supported by recent
studies that provided evidence for an altered vitamin D
gene signature in CD4 + T lymphocytes in patients with
severe COVID-19. Chauss and colleagues [99] demon-
strated that severe COVID-19 may result from a dys-
function of type I immune response, that involves the
vitamin D receptor (VDR) signaling. Similarly, it is inter-
esting to observe how individuals with African descent,
homozygous for the G1 or G2 variant of apolipoprotein
L1 (APOL1), have an increased risk of acute kidney dis-
ease compared to subjects with low-risk variants [100]. A
recent study revealed an association of phenotype sever-
ity and polymorphisms of the MBL2 gene, which encodes
a mannose-binding lectin (MBL) secreted by the liver
and involved in innate immune defense [101]. Innate
immunity is our immune system’s first line of defense
and plays a central role in SARS-CoV-2 infection [102].
Although studied for over 100years, only in recent years
has significant progress has been achieved, largely due to
the genetic dissection of innate immune pathways [103].
Several clinical and immunological studies have shown
that type I interferons (IFN-I) play critical roles in the
control and pathogenesis of COVID-19 [81, 104107].
is notion is supported by extensive sequencing of
numerous patients with severe forms of COVID-19 that
identified pathogenic mutations in genes encoding active
proteins in the interferon circuit [81]. e characteriza-
tion of autoantibodies capable of neutralizing IFN-I in
10–15% of severe patients allows us to state that COVID-
19 can be defined as an interferonopathy [108].
Identifying susceptibility alleles in COVID-19 is impor-
tant in order to improve predictive testing and stratify
different subgroups of SARS-CoV-2 positive subjects,
which can be treated in a personalized way. However, it is
possible that in a complex multifactorial and multigenic
disease, such as COVID-19, several genetic, epigenetic
and socio-demographic factors are modulating the phe-
notypic manifestation, thus complicating the analysis of
genotype–phenotype correlations [32].
Interestingly, the CHGE Consortium (Covid Human
Genetic Effort, https:// www. covid hge. com/ about) initi-
ated a study to enroll individuals (referred to as “resist-
ant”) who were not infected with SARS-CoV-2 despite
repeated 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 [81, 106, 108111]. It is conceivable that
these subjects carry monogenic variations that make
them naturally resistant to virus entry, or much more
active in eliminating the virus by activating appropriate
defense mechanisms such as the genes of the interferon
circuit. Interestingly, a splice variant of OAS1 gene, which
appears to have a protective effect, has been identified
frequently in people of African ancestry [112]. OAS1
encodes for an enzyme catalyzing the synthesis of short
polyadenylates, which activate ribonuclease L that in turn
degrades intracellular double-stranded RNA and triggers
several other antiviral mechanisms [113].
Using trans-ancestry fine-mapping approaches,
Huffman et al. [114] recently demonstrated that the
rs10774671-G splice variant determines the length of the
protein encoded by the gene OAS1, which results in an
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 12
Biancolellaetal. Human Genomics (2022) 16:19
enzyme more effective at breaking down SARS-CoV-2.
It is important to clarify that the genes of susceptibility
to pathogens, although of biological and genetic interest,
cannot in any way confer a sort of “natural immunity” to
infection at an individual level and cannot replace the
important protective role offered by vaccines.
Certainly several “resistance” alleles of genes involved
in the different pathways activated by the infection of
SARS-CoV-2 will be identified in the next months [80, 95,
112]. However, until it is possible to develop polygenic
scores programs which must then be validated on large
numbers, it is unlikely that they can be used to identify
resistant subjects and direct them to selective and spe-
cific therapeutic treatments. ese studies have made it
possible to elucidate many aspects of the pathogenesis of
COVID-19 and have provided many biological responses
to the pathogen-host relationship that could prove
important in other viral infections [81]. In this regard, it
seems interesting to report a recent study that correlates
the loss of smell or taste, very frequent in COVID-19, to
variants of the UGT2A1 and UGT2A2 genes expressed in
the olfactory neuroepithelium, which lines the posterior
nasal cavity, and is exposed to a wide range of odorants
and compounds present in the air [115].
e locations of investigated genes of interest are
resumed in Fig.2 [5, 116].
Characteristics ofavailable vaccines
While witnessing an exponential progress in studies
aimed at understanding genetic and molecular mecha-
nisms, we see their direct application in the tools which
are currently the best candidates to lead us out of this
pandemic: vaccines.
Since the beginning, several critical issues emerged
which led to base the development of vaccines on safety,
immunogenicity, durability of the immunity, dosing
schedule, technological platform and ease of manufac-
ture and transport.
Despite a widespread mistrust about safety and speed
of production, nowadays, we can benefit of two types of
vaccines against SARS-CoV-2 and of a growing number
of data that support their efficacy and safety. Two mes-
senger RNA (mRNA) (BNT162b2 and mRNA-1273)
and two viral vector (ChAdOx1 nCoV-19 AZD1222 and
MRPS21
DPP4
IFIH1
ITGA4
SCN5A
SLC6A20
LZTFL1
GYG1
UGT2A1
UGT2A2
TLR3
ERAP2
HLA
complex
NOTCH4
CCHCR1
TREM1
FOXP4
BRF2
WWP1
TMEM65
IFNB1
C9orf72
EXOSC2
ABO
MBL2
SFTPD
UNC93B1
IFITM3
IRF7
MUC5B
VDR
KLRC2
TBK1
LOC643339
EEA1
OAS1
NEDD4
FURIN
ACE
MAPT
CCL5
ALOXE3
APOE
PLEKHA4
IRF3
NR1H2
DPP9
TICAM1
ICAM1
RAVER1
TYK2
IFNAR2
IFNAR1
KCNE1
TMPRSS2
APOL1
TNFRSF13C
TLR7
ACE2
Fig. 2 Chromosome ideogram representing the location of genes of interest investigated for a role in defining susceptibility to SARS‑CoV‑2
infection (generated by ensembl.org [116])
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 12
Biancolellaetal. Human Genomics (2022) 16:19
Ad26.COV2.S) COVID-19 vaccines were developed
[117]. A third type, protein subunit vaccines (NVX-
CoV2373), has been approved by EMA, Indonesia,
Australia and South Korea, while is still lacking a full
approval by the FDA.
Findings show that the Pfizer/BioNTech BNT162B2
vaccine is safe, with very rare incidence of myocardi-
tis and swelling of the lymph nodes while coronavirus
infection is associated with numerous serious adverse
events such as increased risk of pericarditis, arrhythmias,
heart attacks, strokes, pulmonary embolism, deep-vein
thrombosis, acute kidney damage, and others [118]. e
BNT162b2 COVID-19 vaccine has been shown to reduce
viral load of breakthrough infections (BTIs), but its effec-
tiveness declines after the third to fourth month [119].
e appearance of new SARS-CoV-2 variants poses
new challenges for the development of vaccination plat-
forms [120]. As a consequence, mRNA booster vaccines
were developed to restore the viral neutralization activ-
ity that wanes after the initial two-dose vaccination, to
maintain protection against emerging variants and to
increase vaccine effectiveness in low immune response
individuals such as elderly or immune suppressed. e
need for multiple doses of the vaccine has sparked new
debates, but evidence shows that vaccination with two
doses of mRNA-1273 (Moderna) and a booster are safe
and effective [121]. Moreover, the effectiveness of a third
BNT162b2 vaccine booster was demonstrated in both
reducing transmission and severe disease [122].
As previously stated, we are now aware that higher age
and comorbidities are risk factors for poor outcomes,
regardless of vaccination status [123, 124].
Among adolescents aged 16–17 years, 2-dose mRNA
vaccine effectiveness increased to 86% a week days after
booster dose and urgent care hospitalizations were sub-
stantially lower during the Omicron period than dur-
ing the B.1.617.2 (Delta) predominant period among
adolescents aged 12–17 years, with no significant pro-
tection 150 days after dose 2 during Omicron pre-
dominance [125]. An increasing number of studies are
focusing on the efficacy of multiple doses in fragile cat-
egories: cancer patients receiving at least two doses of
COVID-19 vaccine show reduced risk of COVID-19
[126]. Despite diffused concerns, it has now been estab-
lished that the BNT162b2 and Ad26.COV2.S vaccines
can be safely administered during the third trimester of
pregnancy, reporting excellent results in terms of immu-
nogenicity [127].
e majority of vaccinated patients who required hos-
pitalization due to COVID-19 were elderly with a high
comorbidity burden thus being unable to develop a
proper immune response following vaccination [128].
CDC recommends that all persons aged 12 years and
older receive a booster dose of COVID-19mRNA vac-
cineat least 5months after completing the primary vac-
cination series (at least 2 months after receiving J&J/
Janssen COVID-19 vaccination) and that adults 50years
and older, and moderately or severely immunocompro-
mised people, if eligible, should receive a second booster
dose at least 4months after the previous one.
Vaccines impact on containing the pandemic escala-
tion is evident [129]. eir effectiveness is clearly dose-
dependent as it is higher after administration of a third
dose compared to a second dose administration; how-
ever, vaccine effectiveness wanes with time [129]. For this
reason, and because of the emerging variants that might
overtake the currently available vaccines, the develop-
ment of new vaccines, including variant-specific ones,
should be encouraged and strongly supported. As of
April 15, 2022, 153 vaccines are listed in clinical devel-
opment and 195 in pre-clinical development (WHO,
https:// www. who. int/ publi catio ns/m/ item/ draft- lands
cape- of- covid- 19- candi date- vacci nes, accessed on April
15, 2022) [130].
Outlook
At the official age of about 3years, SARS-CoV-2 is no
longer a baby, but it has proven a rather vicious toddler.
Although ascribing intentions to a virus is naively anthro-
pomorphic, we have to admit that it has managed to cut
short the lives of many fellow-human beings, to change
our ways of relating to each other, to subvert economies,
to shift the priorities of pharmaceutical industry and of
regulatory bodies. With respect to biomedical and pub-
lic health research, as of May29, there are 262,077,248
papers on COVID-19 listed in PubMed, but later tonight
there will be more; and when we wish to discuss science,
we pretend that meeting on line is just as good as being
together in a seminar room – but it is not true.
From the evolutionary point of view, it has been a long-
held tenet of parasitology that for a parasite what is at a
premium is not to kill the host; rather, to have the host
producing the maximum amount of parasite progeny.
SARS-CoV-2 is a perfect illustration. e people infected
have been at least half a billion: Mortality has been there-
fore high in absolute numbers, but at least 98% infected
people have survived and have helped to spread the virus.
Since the beginning of the pandemic, there have been
thousands of mutations in the virus, most of them bio-
logically neutral; at the moment, the predominant Omi-
cron variant seems to be a compromise between high
infectivity and relatively low mortality: Seen from the
vantage point of the virus, the compromise is good, but
not necessarily optimal as yet.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 12
Biancolellaetal. Human Genomics (2022) 16:19
e wealth of studies that have explored the genetic
variation in host susceptibility to SARS-CoV-2 is impres-
sive. ere seem to be genetic factors that affect the
probability of contracting the infection; and many stud-
ies have naturally preferred to focus on genetic factors
that may allow the disease to become severe and life-
threatening. We must admit that we have not yet hit the
jackpot: Nobody has found a human genotype that bars
infection, as homozygosity for the delta 32 allele of the
CCR5 gene does for HIV [131]. Perhaps this is not alto-
gether surprising: For an infectious agent to select a pro-
tective gene, it requires exposure to be continuous over
the host’s many generations [132], as has been the case
for P falciparum and the hemoglobin S gene [133]. If the
postulated jump of SARS-CoV-2 into the human species
has been very recent, there has been no time for selection
of people having an originally rare mutant gene.
In last year’s update [13], we did briefly speculate about
the apparently low numbers of COVID-19 cases in tropi-
cal Africa, and according to the WHO dashboard, this is
still the case. In the two countries, we know best, there
have been, officially, in Nigeria (population over 200 mil-
lion) 119,322 cases and 1926 deaths to date; in Tanzania
(population around 60 million), which for over a year has
not reported cases, there are now on record 33,726 cases
and 800 deaths. We do not yet know to what extent these
low figures are due to underestimation; but from recent
visits, we know that health workers are vaccinated. As for
people in general, what we have learned to my surprise is
that vaccine availability is not currently a limiting factor:
Even when vaccination is free, uptake is quite low.
Anti-COVID-19 vaccination has been a success of
technology and, in many countries, of public health cam-
paigns. Some of us never thought that an effective vac-
cine could be designed, produced and field-tested within
one year: but we have to stand corrected. In retrospect,
the notion of using RNA to immunize against an RNA
virus seems straightforward: Ugur Sahin and Ozlem
Tureci deserve full scientific credit (in addition to many
millions in revenue) for what they have achieved. With
most previous vaccines, peptides derived from the organ-
ism or from the purified protein injected are presented to
T cells by Antigen Presenting Cells (APC). In this case,
instead, nanoparticles that encapsulate the portion of the
viral RNA that encodes the spike protein are endocytosed
by the APCs, whose protein synthetic machinery is taken
over to translate the RNA into the spike protein, peptides
from which are then presented to T cells. A key to the
ultra-fast development of the BioNTech vaccine has been
this clever approach: Since it was unprecedented, nobody
was entitled to predict (i) How it would work in prac-
tice, (ii) What would be the duration of immunity. With
respect to (i) e results have been spectacular; with
respect to (ii) One had to find out the answer empirically,
and it turned out that, even after two doses, the dura-
tion is of the order of months, not years. Only the oldest
among us can remember that when mRNA was discov-
ered, one of its defining properties was a short life span
[134]: It seems not far-fetched to hypothesize that the
takeover of the APC’s ribosomes by SARS-CoV-2 mRNA
is short-lived, and this may be at least one reason why
immunity does not last very long.
Unlike the COVID-19 epidemic, the epidemic of no-
Vax does not lend itself to mathematical analysis: It is
not in the realm of biological science, but rather in the
realm of psycho-patho-sociology [135]. We have all
learnt that evidence-based reasoning does not make a
dent in the hard-core no-Vax. Confrontation fails, and
an approach based on lateral thinking may be better.
Perhaps we should take this on as an intellectual chal-
lenge: If we can largely prevent severe COVID-19 by
immunization, if we can find the molecular basis of
many diseases, if in many cases we can cure leukemia,
we should be able to also address the no-Vax problem.
Abbreviations
ACE2: Angiotensin‑converting enzyme 2; APC: Antigen presenting cells;
APOL1: Apolipoprotein L1; BBS: Bardet‑Biedl syndrome; CCL5: CC‑chemokine
ligand 5; CDC: Centers for disease control and prevention; CHGE: Covid human
genetic effort; COVID‑19: Coronavirus disease 2019; CSSE: Center for systems
and software engineering; CT: Computed tomography; DPP4: Dipeptidyl
peptidase 4; EEA1: Early endosomal antigen 1; EMA: European medicines
agency; FDA: Food and drug administration; FDG: Fluorodeoxyglucose; GWAS:
Genome‑wide association study; H1N1: Hemagglutinin type 1 and neurami‑
nidase type 1; HLA: Human leukocyte antigens; ICAM1: Inter‑cellular adhesion
molecule 1; IFN: Interferon; IFNB1: Interferon beta 1; LZTFL1: Leucine zipper
transcription factor like 1; MBL: Mannose‑binding lectin; MDA5: Melanoma
differentiation‑associated rotein 5; MUC5B: Mucin protein 5B; NEDD4: Neural
precursor cell expressed developmentally down‑regulated protein 4; NFE2:
Nuclear factor erythroid 2; NRF2: Nuclear factor erythroid 2 (NFE2)‑related
factor 2; OAS1: 2’‑5’‑Oligoadenylate synthetase 1; PH: Public Health; PheWAS:
Phenome‑wide association study; RAVER1: Ribonucleoprotein PTB‑binding
1; SARS‑CoV‑2: Severe acute respiratory syndrome coronavirus‑2; SFTPD:
Surfactant protein D; SNP: Single nucleotide polymorphism; TMPRSS2:
Transmembrane serine protease 2; TNF: Tumor necrosis factor; UGT2A: UDP‑
glucuronosyltransferase 2A; VDR: Vitamin D receptor; VOC: Variant of concern;
WHO: World health organization; WWP1: WW domain containing E3 ubiquitin
protein ligase 1.
Acknowledgements
We thank Mira Nagel for permission to reproduce the cartoon presented in
Figure 1. We are grateful for the assistance given by Dr. Francesca Pisanu for
her help with the editing and organization of the manuscript.
Author contributions
JKVR conceived the manuscript, wrote the historical and transition part and
edited the paper. JLW edited the manuscript. VLC wrote the introduction and
clinical manifestations sections, conceived and edited Fig. 2, and revised the
manuscript. GN and MB wrote the genetic susceptibility part, conceived Fig. 2
and revised the paper. LL wrote the outlook section and performed review
and revision of the paper. RMS wrote the characteristics of available vaccines
paragraph. All authors read and approved the final version of the manuscript.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 12
Biancolellaetal. Human Genomics (2022) 16:19
Funding
This study was also supported in part by a grant of Regione Lazio (Italy, Pro‑
getti di Gruppi di Ricerca 2020 A0375‑2020–36663 GecoBiomark) and Rome
Foundation (Italy, Prot 317A/I) to GN.
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
Competing interests
The authors declare that they have no competing interests.
Author details
1 Department of Biology, Tor Vergata University of Rome, Rome, Italy.
2 Department of Biomedicine and Prevention, Tor Vergata University of Rome,
00133 Rome, Italy. 3 Sheba Medical Center, Pediatric Hemato‑Oncology,
Edmond and Lilly Safra Children’s Hospital, Tel Hashomer 2 Sheba Road,
52621 Ramat Gan, Israel. 4 College of Public Health, Medical and Veterinary
Sciences, James Cook University, Smithfield, QLD 4878, Australia. 5 Depart‑
ment of Haematology and Blood Transfusion, Muhimbili University of Health
and Allied Sciences, Dar es Salaam, Tanzania. 6 University of Florence, Florence,
Italy. 7 IRCCS Neuromed, Pozzilli, Isernia, Italy. 8 Department of Pharmacology,
School of Medicine, University of Nevada, Reno, NV, USA. 9 Australian Institute
of Tropical Health and Medicine, James Cook University, Smithfield, QLD 4878,
Australia. 10 Department of Biomedicine and Prevention, School of Medicine
and Surgery, Via Montpellier 1, 00133 Rome, Italy.
Received: 22 March 2022 Accepted: 26 April 2022
References
1. Cucinotta D, Vanelli M. WHO declares COVID‑19 a pandemic. Acta
Biomed. 2020;91(1):157–60.
2. National Center for Immunization and Respiratory Diseases (NCIRD),
Division of Viral Diseases. CDC COVID‑19 Science Briefs. Atlanta (GA):
Centers for Disease Control and Prevention (US); 2020–. Science Brief:
Omicron (B.1.1.529) Variant. 2021 Dec 2.
3. Queen D. Another year another variant: COVID 3.0‑omicron. Int Wound
J. 2022;19(1):5. https:// doi. org/ 10. 1111/ iwj. 13739.
4. Abdullah F, Myers J, Basu D, Tintinger G, Ueckermann V, Mathebula M,
et al. Decreased severity of disease during the first global omicron vari‑
ant covid‑19 outbreak in a large hospital in tshwane, south africa. Int
J Infect Dis. 2022;116:38–42. https:// doi. org/ 10. 1016/j. ijid. 2021. 12. 357
(Epub 2021 Dec 28).
5. Colona VL, Vasiliou V, Watt J, Novelli G, Reichardt JKV. Update on human
genetic susceptibility to COVID‑19: susceptibility to virus and response.
Hum Genom. 2021;15(1):57. https:// doi. org/ 10. 1186/ s40246‑ 021‑ 00356‑
x. Errat um. In: HumGe nomics. 2021S ep18; 15(1): 59.
6. Dong E, Du H, Gardner L. An interactive web‑based dashboard to track
COVID‑19 in real time. Lancet Infect Dis. 2020; 20(5):533–534. https://
doi. org/ 10. 1016/ S1473‑ 3099(20) 30120‑1. Epub 2020 Feb 19. Erratum in:
Lancet Infect Dis. 2020; 20(9):e215.
7. O’Toole Á, Hill V, Pybus OG, Watts A, Bogoch II, Khan K, et al. Track‑
ing the international spread of SARS‑CoV‑2 lineages B.1.1.7 and
B.1.351/501Y‑V2 with grinch. Wellcome Open Res. 2021;6:121.
8. CDC. Centers for disease control and prevention. Covid.cdc.gov “Covid.
cdc.gov “.2022; Retrieved 19/04, 2022, from https:// covid. cdc. gov/ covid‑
data‑ track er.
9. Altarawneh HN, Chemaitelly H, Hasan MR, Ayoub HH, Qassim S,
AlMukdad S, et al. Protection against the Omicron variant from previous
SARS‑CoV‑2 Infection. N Engl J Med. 2022. https:// doi. org/ 10. 1056/
NEJMc 22001 33.
10. Chaguza C, Coppi A, Earnest R, Ferguson D, Kerantzas N, Warner F, et al.
Rapid emergence of SARS‑CoV‑2 Omicron variant is associated with an
infection advantage over Delta in vaccinated persons. Med (N Y). 2022.
https:// doi. org/ 10. 1016/j. medj. 2022. 03. 010.
11. Pulliam JRC, van Schalkwyk C, Govender N, von Gottberg A, Cohen C,
Groome MJ, et al. Increased risk of SARS‑CoV‑2 reinfection associated
with emergence of Omicron in South Africa. Science. 2022. https:// doi.
org/ 10. 1126/ scien ce. abn49 47.
12. COVID‑19 Reinfection Data (2022). Retrieved 19/04; 2022, from https://
coron avirus. health. ny. gov/ covid‑ 19‑ reinf ection‑ data.
13. Novelli G, Biancolella M, Mehrian‑Shai R, Colona VL, Brito AF, Gru‑
baugh ND, et al. COVID‑19 one year into the pandemic: from genetics
and genomics to therapy, vaccination, and policy. Hum Genom.
2021;15(1):27. https:// doi. org/ 10. 1186/ s40246‑ 021‑ 00326‑3.
14. Muhsen K, Maimon N, Mizrahi A, Varticovschi B, Bodenheimer O,
Gelbshtein U, et al. Effects of BNT162b2 Covid‑19 vaccine booster in
long‑term care facilities in Israel. N Engl J Med. 2022;386(4):399–401.
https:// doi. org/ 10. 1056/ NEJMc 21173 85 (Epub 2021 Dec 22).
15. Tenforde MW, Self WH, Adams K, Gaglani M, Ginde AA, McNeal T, et al.
Influenza and other viruses in the acutely Ill (IVY) network association
between mRNA vaccination and COVID‑19 hospitalization and disease
severity. JAMA. 2021;326(20):2043–54. https:// doi. org/ 10. 1001/ jama.
2021. 19499.
16. Tregoning JS, Flight KE, Higham SL, Wang Z, Pierce BF. Progress of the
COVID‑19 vaccine effort: viruses, vaccines and variants versus efficacy,
effectiveness and escape. Nat Rev Immunol. 2021;21(10):626–36.
https:// doi. org/ 10. 1038/ s41577‑ 021‑ 00592‑1 (Epub 2021 Aug 9).
17. Collie S, Champion J, Moultrie H, Bekker LG, Gray G. Effectiveness of
BNT162b2 vaccine against omicron variant in South Africa. N Engl J
Med. 2022;386(5):494–6. https:// doi. org/ 10. 1056/ NEJMc 21192 70 (Epub
2021 Dec 29).
18. Davis JT, Chinazzi M, Perra N, Mu K, Pastore Y, Piontti A, et al. Cryptic
transmission of SARS‑CoV‑2 and the first COVID‑19 wave. Nature.
2021;600(7887):127–32. https:// doi. org/ 10. 1038/ s41586‑ 021‑ 04130‑w.
19. Green MS. Did the hesitancy in declaring COVID‑19 a pandemic reflect
a need to redefine the term? Lancet. 2020;395(10229):1034–5. https://
doi. org/ 10. 1016/ S0140‑ 6736(20) 30630‑9 (Epub 2020 Mar 13).
20. WHO. World Health Organization. Who.int “who.int”. 2022; Retrieved
19/04, 2022, from https:// www. who. int/ news/ item/ 13‑ 10‑ 2021‑ who‑
annou nces‑ propo sed‑ membe rs‑ of‑ its‑ scien tific‑ advis ory‑ group‑ for‑
the‑ origi ns‑ of‑ novel‑ patho gens‑ (sago).
21. COVID is here to stay. countries must decide how to adapt. Nature.
2022;601(7892):165. https:// doi. org/ 10. 1038/ d41586‑ 022‑ 00057‑y.
22. Adam D. Will Omicron end the pandemic? Here’s What Experts
Say Nature. 2022;602(7895):20–1. https:// doi. org/ 10. 1038/
d41586‑ 022‑ 00210‑7.
23. Happi C T, Nkengasong JN. Two years of COVID‑19 in Africa: lessons for
the world. Nature. 2022.
24. Ser vick K. Is it time to live with COVID‑19? Some scientists warn of
‘endemic delusion. Science. 2022.
25. Hale VL, Dennis PM, McBride DS, Nolting JM, Madden C, Huey D,
et al. SARS‑CoV‑2 infection in free‑ranging white‑tailed deer. Nature.
2022;602(7897):481–6. https:// doi. org/ 10. 1038/ s41586‑ 021‑ 04353‑x
(Epub 2021 Dec 23).
26. Kumar A, Narayan RK, Prasoon P, Kumari C, K aur G, Kumar S, et al.
COVID‑19 mechanisms in the human body‑what we know so far. Front
Immunol. 2021;1(12):693938. https:// doi. org/ 10. 3389/ fimmu. 2021.
693938.
27. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al. Epidemio‑
logical and clinical characteristics of 99 cases of 2019 novel coro‑
navirus pneumonia in Wuhan, China: a descriptive study. Lancet.
2020;395(10223):507–13. https:// doi. org/ 10. 1016/ S0140‑ 6736(20)
30211‑7 (Epub 2020 Jan 30).
28. 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):610–4. https:// doi. org/ 10.
1111/ tan. 14047 (Epub 2020 Sep 3).
29. Chao JY, Derespina KR, Herold BC, Goldman DL, Aldrich M, Weingarten
J, et al. Clinical characteristics and outcomes of hospitalized and criti‑
cally Ill children and adolescents with coronavirus disease 2019 at a
tertiary care medical center in New York City. J Pediatr. 2020;223:14‑19.
e2. https:// doi. org/ 10. 1016/j. jpeds. 2020. 05. 006 (Epub 2020 May 11).
30. Harrison AG, Lin T, Wang P. Mechanisms of SARS‑CoV‑2 transmission
and pathogenesis. Trends Immunol. 2020;41(12):1100–15. https:// doi.
org/ 10. 1016/j. it. 2020. 10. 004 (Epub 2020 Oct 14).
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 12
Biancolellaetal. Human Genomics (2022) 16:19
31. Zhang Q, Bastard P, Human Genetic Effort COVID, Cobat A, Casanova JL.
Human genetic and immunological determinants of critical COVID‑19
pneumonia. Nature. 2022. https:// doi. org/ 10. 1038/ s41586‑ 022‑ 04447‑0.
32. Colona VL, Biancolella M, Novelli A, Novelli G. Will GWAS eventually
allow the identification of genomic biomarkers for COVID‑19 severity
and mortality? J Clin Invest. 2021;131(23):e155011. https:// doi. org/ 10.
1172/ JCI15 5011.
33. Huang L, Yao Q, Gu X, Wang Q, Ren L, Wang Y, et al. 1‑year outcomes in
hospital survivors with COVID‑19: a longitudinal cohort study. Lancet.
2021;398(10302):747–58. https:// doi. org/ 10. 1016/ S0140‑ 6736(21)
01755‑4.
34. Sudre CH, Murray B, Varsavsky T, Graham MS, Penfold RS, Bowyer
RC, et al. Attributes and predictors of long COVID. Nat Med. 2021;
27(4):626–631. https:// doi. org/ 10. 1038/ s41591‑ 021‑ 01292‑y. Epub 2021
Mar 10. Erratum in: Nat Med. 2021; 27(6):1116.
35. Visco V, Vitale C, Rispoli A, Izzo C, Virtuoso N, Ferruzzi GJ, et al. Post‑
COVID‑19 Syndrome: Involvement and Interactions between respira‑
tory, cardiovascular and nervous systems. J Clin Med. 2022;11(3):524.
https:// doi. org/ 10. 3390/ jcm11 030524.
36. Fraser E. Persistent pulmonary disease after acute covid‑19. BMJ.
2021;21(373):n1565. https:// doi. org/ 10. 1136/ bmj. n1565.
37. Grist JT, Chen M, Collier GJ, Raman B, Abueid G, McIntyre A, et al. Hyper‑
polarized 129Xe MRI abnormalities in dyspneic patients 3 months after
COVID‑19 pneumonia: preliminary results. Radiology. 2021;301(1):E353–
60. https:// doi. org/ 10. 1148/ radiol. 20212 10033 (Epub 2021 May 25).
38. Torres‑Castro R, Vasconcello‑Castillo L, Alsina‑Restoy X, Solis‑Navarro L,
Burgos F, Puppo H, et al. Respiratory function in patients post‑infection
by COVID‑19: a systematic review and meta‑analysis. Pulmonology.
2021;27(4):328–37. https:// doi. org/ 10. 1016/j. pulmoe. 2020. 10. 013.
39. Nalbandian A, Sehgal K, Gupta A, Madhavan MV, McGroder C, Stevens
JS, et al. Post‑acute COVID‑19 syndrome. Nat Med. 2021;27(4):601–15.
https:// doi. org/ 10. 1038/ s41591‑ 021‑ 01283‑z (Epub 2021 Mar 22).
40. Burgess LC, Venugopalan L, Badger J, Street T, Alon G, Jarvis JC, et al.
Effect of neuromuscular electrical stimulation on the recovery of
people with COVID‑19 admitted to the intensive care unit: a narrative
review. J Rehabil Med. 2021;53(3):jrm00164. https:// doi. org/ 10. 2340/
16501 977‑ 2805.
41. Fernández‑de‑Las‑Peñas C, Rodríguez‑Jiménez J, Fuensalida‑Novo S,
Palacios‑Ceña M, Gómez‑Mayordomo V, Florencio LL, et al. Myalgia as a
symptom at hospital admission by severe acute respiratory syndrome
coronavirus 2 infection is associated with persistent musculoskeletal
pain as long‑term post‑COVID sequelae: a case‑control study. Pain.
2021;162(12):2832–40. https:// doi. org/ 10. 1097/j. pain. 00000 00000
002306.
42. Farr E, Wolfe AR, Deshmukh S, Rydberg L, Soriano R, Walter JM, et al.
Diaphragm dysfunction in severe COVID‑19 as determined by neuro‑
muscular ultrasound. Ann Clin Transl Neurol. 2021;8(8):1745–9. https://
doi. org/ 10. 1002/ acn3. 51416 (Epub 2021 Jul 11).
43. Rodriguez B, Nansoz S, Cameron DR, Z’Graggen WJ. Is myopathy part of
long‑Covid? Clin Neurophysiol. 2021;132(6):1241–2. https:// doi. org/ 10.
1016/j. clinph. 2021. 03. 008 (Epub 2021 Mar 26).
44. Silva Andrade B, Siqueira S, de Assis Soares WR, de Souza RF, Santos NO,
Dos Santos FA, et al. Long‑COVID and post‑COVID health complications:
an up‑to‑date review on clinical conditions and their possible molecu‑
lar mechanisms. Viruses. 2021;13(4):700. https:// doi. org/ 10. 3390/ v1304
0700.
45. Stöbe S, Richter S, Seige M, Stehr S, Laufs U, Hagendorff A. Echocardio‑
graphic characteristics of patients with SARS‑CoV‑2 infection. Clin Res
Cardiol. 2020;109(12):1549–66. https:// doi. org/ 10. 1007/ s00392‑ 020‑
01727‑5 (Epub 2020 Aug 14).
46. Taquet M, Geddes JR, Husain M, Luciano S, Harrison PJ. 6‑month neu‑
rological and psychiatric outcomes in 236 379 survivors of COVID‑19:
a retrospective cohort study using electronic health records. Lancet
Psychiatry. 2021;8(5):416–27. https:// doi. org/ 10. 1016/ S2215‑ 0366(21)
00084‑5 (Epub 2021 Apr 6).
47. Garcia‑Azorin D, Layos‑Romero A, Porta‑Etessam J, Membrilla JA,
Caronna E, Gonzalez‑Martinez A, et al. Post‑COVID‑19 persistent head‑
ache: a multicentric 9‑months follow‑up study of 905 patients. Cephala‑
lgia. 2022;15:3331024211068074. https:// doi. org/ 10. 1177/ 03331 02421
10680 74.
48. Xydakis MS, Albers MW, Holbrook EH, Lyon DM, Shih RY, Frasnelli JA,
et al. Post‑viral effects of COVID‑19 in the olfactory system and their
implications. Lancet Neurol. 2021;20(9):753–61. https:// doi. org/ 10. 1016/
S1474‑ 4422(21) 00182‑4 (Epub 2021 Jul 30).
49. Mazzatenta A, Montagnini C, Brasacchio A, Sartucci F, Neri G. Electro‑
physiological and olfactometric evaluation of long‑term COVID‑19.
Physiol Rep. 2021;9(18):e14992.
50. Guedj E, Lazarini F, Morbelli S, Ceccaldi M, Hautefort C, Kas A, et al. Long
COVID and the brain network of Proust’s madeleine: targeting the olfac‑
tory pathway. Clin Microbiol Infect. 2021;27(9):1196–8. https:// doi. org/
10. 1016/j. cmi. 2021. 05. 015 (Epub 2021 May 17).
51. Graham EL, Clark JR, Orban ZS, Lim PH, Szymanski AL, Taylor C, et al. Per
sistent neurologic symptoms and cognitive dysfunction in non‑hospi‑
talized Covid‑19 “long haulers.” Ann Clin Transl Neurol. 2021;8(5):1073–
85. https:// doi. org/ 10. 1002/ acn3. 51350 (Epub 2021 Mar 30).
52. MacIntosh BJ, Ji X, Chen JJ, Gilboa A, Roudaia E, Sekuler AB, Gao F, et al.
Brain structure and function in people recovering from COVID‑19 after
hospital discharge or self‑isolation: a longitudinal observational study
protocol. CMAJ Open. 2021;9(4):E1114–9. https:// doi. org/ 10. 9778/
cmajo. 20210 023.
53. Lorkiewicz P, Waszkiewicz N. Biomarkers of Post‑COVID depression. J
Clin Med. 2021;10(18):4142. https:// doi. org/ 10. 3390/ jcm10 184142.
54. Raahimi MM, Kane A, Moore CE, Alareed AW. Late onset of Guillain‑
Barré syndrome following SARS‑CoV‑2 infection: part of “long COVID‑19
syndrome”? BMJ Case Rep. 2021;14(1):e240178. https:// doi. org/ 10. 1136/
bcr‑ 2020‑ 240178.
55. Kakumoto T, Kobayashi S, Yuuki H, Kainaga M, Shirota Y, Hamada M,
et al. Cranial nerve involvement and dysautonomia in post‑COVID‑19
Guillain‑Barré syndrome. Intern Med. 2021;60(21):3477–80. https:// doi.
org/ 10. 2169/ inter nalme dicine. 7355‑ 21 (Epub 2021 Aug 24).
56. Reyes‑Bueno JA, García‑Trujillo L, Urbaneja P, Ciano‑Petersen NL,
Postigo‑Pozo MJ, Martínez‑Tomás C, et al. Miller‑fisher syndrome after
SARS‑CoV‑2 infection. Eur J Neurol. 2020;27(9):1759–61. https:// doi. org/
10. 1111/ ene. 14383.
57. Salihefendic N, Zildzic M, Huseinagic H. Ischemic vasculitis as a cause
of brain disorder’s in patients with long Covid: case report. Med Arch.
2021;75(6):471–4. https:// doi. org/ 10. 5455/ medarh. 2021. 75. 471‑ 474.
58. Barizien N, Le Guen M, Russel S, Touche P, Huang F, Vallée A. Clinical
characterization of dysautonomia in long COVID‑19 patients. Sci Rep.
2021;11(1):14042. https:// doi. org/ 10. 1038/ s41598‑ 021‑ 93546‑5.
59. Al‑Kuraishy HM, Al‑Gareeb AI, Qusti S, Alshammari EM, Gyebi GA, Batiha
GE. Covid‑19‑induced dysautonomia: a menace of sympathetic storm.
ASN Neuro. 2021;13:17590914211057636. https:// doi. org/ 10. 1177/
17590 91421 10576 35.
60. Bo HX, Li W, Yang Y, Wang Y, Zhang Q, Cheung T, et al. Posttraumatic
stress symptoms and attitude toward crisis mental health services
among clinically stable patients with COVID‑19 in China. Psychol Med.
2021;51(6):1052–3. https:// doi. org/ 10. 1017/ S0033 29172 00009 99 (Epub
2020 Mar 27).
61. Gilio L, Galifi G, Centonze D, Stampanoni BM. Case report: overlap
between long covid and functional neurological disorders. Front Neu‑
rol. 2022;28(12):811276. https:// doi. org/ 10. 3389/ fneur. 2021. 811276.
62. Wu Y, Xu X, Chen Z, Duan J, Hashimoto K, Yang L, et al. Nervous system
involvement after infection with COVID‑19 and other coronaviruses.
Brain Behav Immun. 2020;87:18–22. https:// doi. org/ 10. 1016/j. bbi. 2020.
03. 031 (Epub 2020 Mar 30).
63. Azizi SA, Azizi SA. Neurological injuries in COVID‑19 patients: direct viral
invasion or a bystander injury after infection of epithelial/endothelial
cells. J Neurovirol. 2020;26(5):631–41. https:// doi. org/ 10. 1007/ s13365‑
020‑ 00903‑7 (Epub 2020 Sep 2).
64. Kanberg N, Simrén J, Edén A, Andersson LM, Nilsson S, Ashton NJ, et al.
Neurochemical signs of astrocytic and neuronal injury in acute COVID‑
19 normalizes during long‑term follow‑up. EBioMedicine. 2021;70:
103512. https:// doi. org/ 10. 1016/j. ebiom. 2021. 103512 (Epub 2021 Jul
29).
65. Carroll E, Neumann H, Aguero‑Rosenfeld ME, Lighter J, Czeisler BM,
Melmed K, et al. Post‑COVID‑19 inflammatory syndrome manifesting as
refractory status epilepticus. Epilepsia. 2020;61(10):e135–9. https:// doi.
org/ 10. 1111/ epi. 16683 (Epub 2020 Sep 18).
66. Zhou Z, Kang H, Li S, Zhao X. Understanding the neurotropic charac‑
teristics of SARS‑CoV‑2: from neurological manifestations of COVID‑19
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 12
Biancolellaetal. Human Genomics (2022) 16:19
to potential neurotropic mechanisms. J Neurol. 2020;267(8):2179–84.
https:// doi. org/ 10. 1007/ s00415‑ 020‑ 09929‑7 (Epub 2020 May 26).
67. Matschke J, Lütgehetmann M, Hagel C, Sperhake JP, Schröder AS, Edler
C, et al. Neuropathology of patients with COVID‑19 in Germany: a post‑
mortem case series. Lancet Neurol. 2020;19(11):919–29. https:// doi. org/
10. 1016/ S1474‑ 4422(20) 30308‑2 (Epub 2020 Oct 5).
68. Douaud G, Lee S, Alfaro‑Almagro F, Arthofer C, Wang C, McCarthy P,
et al. SARS‑CoV‑2 is associated with changes in brain structure in UK
Biobank. Nature. 2022. https:// doi. org/ 10. 1038/ s41586‑ 022‑ 04569‑5.
69. Nolen LT, Mukerji SS, Mejia NI. Post‑acute neurological consequences
of COVID‑19: an unequal burden. Nat Med. 2022;28(1):20–3. https:// doi.
org/ 10. 1038/ s41591‑ 021‑ 01647‑5.
70. The Lancet Neurology. Long COVID: understanding the neurological
effects. Lancet Neurol. 2021;20(4):247. https:// doi. org/ 10. 1016/ S1474‑
4422(21) 00059‑4.
71. Jarrott B, Head R, Pringle KG, Lumbers ER, Martin JH. “LONG COVID”‑A
hypothesis for understanding the biological basis and pharmacological
treatment strategy. Pharmacol Res Perspect. 2022;10(1):e00911. https://
doi. org/ 10. 1002/ prp2. 911.
72. Petrillo S, Piermarini E, Pastore A, Vasco G, Schirinzi T, Carrozzo R, et al.
Nrf2‑inducers counteract neurodegeneration in frataxin‑silenced motor
neurons: disclosing new therapeutic targets for Friedreich’s ataxia. Int J
Mol Sci. 2017;18(10):2173. https:// doi. org/ 10. 3390/ ijms1 81021 73.
73. Petrillo S, Schirinzi T, Di Lazzaro G, D’Amico J, Colona VL, Bertini E, et al.
Systemic activation of Nrf2 pathway in Parkinson’s disease. Mov Disord.
2020;35(1):180–4. https:// doi. org/ 10. 1002/ mds. 27878 (Epub 2019 Nov
4).
74. Satoh T, Trudler D, Oh CK, Lipton SA. Potential therapeutic use of the
rosemary diterpene carnosic acid for alzheimer’s disease, parkinson’s
disease, and long‑COVID through NRF2 activation to counteract the
NLRP3 inflammasome. Antioxidants (Basel). 2022;11(1):124. https:// doi.
org/ 10. 3390/ antio x1101 0124.
75. Paul BD, Lemle MD, Komaroff AL, Snyder SH. Redox imbalance links
COVID‑19 and myalgic encephalomyelitis/chronic fatigue syndrome.
Proc Natl Acad Sci U S A. 2021;118(34):e2024358118. https:// doi. org/ 10.
1073/ pnas. 20243 58118.
76. Rovere Querini P, De Lorenzo R, Conte C, Brioni E, Lanzani C, Yacoub
MR, et al. Post‑COVID‑19 follow‑up clinic: depicting chronicity of a new
disease. Acta Biomed. 2020;91(9‑S):22–8.
77. Kell DB, Laubscher GJ, Pretorius E. A central role for amyloid fibrin
microclots in long COVID/PASC: origins and therapeutic implications.
Biochem J. 2022;479(4):537–59. https:// doi. org/ 10. 1042/ BCJ20 220016.
78. Casanova JL, Abel L. Mechanisms of viral inflammation and disease in
humans. Science. 2021;374(6571):1080–6. https:// doi. org/ 10. 1126/ scien
ce. abj79 65.
79. Anastassopoulou C, Gkizarioti Z, Patrinos GP, Tsakris A. Human genetic
factors associated with susceptibility to SARS‑CoV‑2 infection and
COVID‑19 disease severity. Hum Genom. 2020;14(1):40. https:// doi. org/
10. 1186/ s40246‑ 020‑ 00290‑4.
80. Andreakos E, Abel L, Vinh DC, Kaja E, Drolet BA, Zhang Q, et al. A global
effort to dissect the human genetic basis of resistance to SARS‑CoV‑2
infection. Nat Immunol. 2021; https:// doi. org/ 10. 1038/ s41590‑ 021‑
01030‑z. Epub 2021 Oct 18. Erratum in: Nat Immunol. 2021 Nov 24.
81. Zhang Q, Bastard P, Effort CHG, Cobat A, Casanova JL. Human genetic
and immunological determinants of critical COVID‑19 pneumonia.
Nature. 2022. https:// doi. org/ 10. 1038/ s41586‑ 022‑ 04447‑0.
82. Fink‑Baldauf IM, Stuart WD, Brewington JJ, Guo M, Maeda Y. CRISPRi
links COVID‑19 GWAS loci to LZTFL1 and RAVER1. EBioMedicine.
2022;75:103806. https:// doi. org/ 10. 1016/j. ebiom. 2021. 103806 (Epub
2022 Jan 6).
83. COVID‑19 Host Genetics Initiative. Mapping the human genetic archi‑
tecture of COVID‑19. Nature. 2021;600(7889):472–477. https:// doi. org/
10. 1038/ s41586‑ 021‑ 03767‑x. Epub 2021 Jul 8.
84. Chamnanphon M, Pongpanich M, Suttichet TB, Jantarabenjakul W,
Torvorapanit P, Putcharoen O, Sodsai P, Phokaew C, Hirankarn N, Chari‑
yavilaskul P, Shotelersuk V. Host genetic factors of COVID‑19 susceptibil‑
ity and disease severity in a Thai population. J Hum Genet. 2022;11:1–7.
https:// doi. org/ 10. 1038/ s10038‑ 021‑ 01009‑6.
85. Sánchez EG, Pérez‑Núñez D, Revilla Y. Mechanisms of entry and endoso‑
mal pathway of African swine fever virus. Vaccines (Basel). 2017;5(4):42.
https:// doi. org/ 10. 3390/ vacci nes50 40042.
86. Regan JA, Abdulrahim JW, Bihlmeyer NA, Haynes C, Kwee LC, Patel MR,
Shah SH. Phenome‑wide association study of severe COVID‑19 genetic
risk variants. J Am Heart Assoc. 2022;11(5):e024004. https:// doi. org/ 10.
1161/ JAHA. 121. 024004.
87. 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;12(4):9. https:// doi. org/ 10. 1186/
1471‑ 2350‑4‑9.
88. Cho JH, Gregersen PK. Genomics and the multifactorial nature of
human autoimmune disease. N Engl J Med. 2011;365(17):1612–23.
https:// doi. org/ 10. 1056/ NEJMr a1100 030.
89. Dutta M, Dutta P, Medhi S, Borkakoty B, Biswas D. Polymorphism of HLA
class I and class II alleles in influenza A(H1N1)pdm09 virus infected
population of Assam Northeast India. J Med Virol. 2018;90(5):854–60.
https:// doi. org/ 10. 1002/ jmv. 25018.
90. Ma Y, Yuan B, Yi J, Zhuang R, Wang J, Zhang Y, et al. The genetic poly‑
morphisms of HLA are strongly correlated with the disease severity
after Hantaan virus infection in the Chinese Han population. Clin Dev
Immunol. 2012;2012:308237. https:// doi. org/ 10. 1155/ 2012/ 308237
(Epub 2012 Oct 8).
91. Migliorini F, Torsiello E, Spiezia F, Oliva F, Tingart M, Maffulli N. Associa‑
tion between HLA genotypes and COVID‑19 susceptibility, severity and
progression: a comprehensive review of the literature. Eur J Med Res.
2021;26(1):84. https:// doi. org/ 10. 1186/ s40001‑ 021‑ 00563‑1.
92. Weiner J, Suwalski P, Holtgrewe M, Rakitko A, Thibeault C, Müller M, et al.
Increased risk of severe clinical course of COVID‑19 in carriers of HLA‑
C*04:01. EClinicalMedicine. 2021;40:101099. https:// doi. org/ 10. 1016/j.
eclinm. 2021. 101099 (Epub 2021 Sep 2).
93. 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/ genes 11091 010.
94. Hoffmann M, Kleine‑Weber H, Schroeder S, Krüger 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):271‑
280.e8. https:// doi. org/ 10. 1016/j. cell. 2020. 02. 052 (Epub 2020 Mar 5).
95. David A, Parkinson N, Peacock TP, Pairo‑Castineira E, Khanna T, Cobat A,
et al. A common TMPRSS2 variant has a protective effect against severe
COVID‑19. Curr Res Transl Med. 2022;70(2):103333. https:// doi. org/ 10.
1016/j. retram. 2022. 103333.
96. Pandey RK, Srivastava A, Singh PP, Chaubey G. Genetic association of
TMPRSS2 rs2070788 polymorphism with COVID‑19 case fatality rate
among Indian populations. Infect Genet Evol. 2022;98:105206. https://
doi. org/ 10. 1016/j. meegid. 2022. 105206 (Epub 2022 Jan 5).
97. Novelli G, Liu J, Biancolella M, Alonzi T, Novelli A, Patten JJ, et al. Inhibi‑
tion 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.
98. Freitas AT, Calhau C, Antunes G, Araújo B, Bandeira M, Barreira S, et al.
Vitamin D‑related polymorphisms and vitamin D levels as risk biomark‑
ers of COVID‑19 disease severity. Sci Rep. 2021;11(1):20837. https:// doi.
org/ 10. 1038/ s41598‑ 021‑ 99952‑z.
99. Chauss D, Freiwald T, McGregor R, Yan B, Wang L, Nova‑Lamperti E, et al.
Autocrine vitamin D signaling switches off pro‑inflammatory programs
of TH1 cells. Nat Immunol. 2022;23(1):62–74. https:// doi. org/ 10. 1038/
s41590‑ 021‑ 01080‑3 (Epub 2021 Nov 11).
100. Hung AM, Shah SC, Bick AG, Yu Z, Chen HC, Hunt CM, et al. APOL1 Risk
variants, acute kidney injury, and death in participants with African
ancestry hospitalized With COVID‑19 from the million veteran program.
JAMA Intern Med. 2022. https:// doi. org/ 10. 1001/ jamai ntern med. 2021.
8538.
101. Stravalaci M, Pagani I, Paraboschi EM, Pedotti M, Doni A, Scavello F, et al.
Recognition and inhibition of SARS‑CoV‑2 by humoral innate immunity
pattern recognition molecules. Nat Immunol. 2022;23(2):275–86.
https:// doi. org/ 10. 1038/ s41590‑ 021‑ 01114‑w (Epub 2022 Jan 31).
102. Schultze JL, Aschenbrenner AC. COVID‑19 and the human innate
immune system. Cell. 2021;184(7):1671–92. https:// doi. org/ 10. 1016/j.
cell. 2021. 02. 029 (Epub 2021 Feb 16).
103. Beutler B. Innate immunity: an overview. Mol Immunol.
2004;40(12):845–59. https:// doi. org/ 10. 1016/j. molimm. 2003. 10. 005.
104. Mantovani S, Daga S, Fallerini C, Baldassarri M, Benetti E, Picchiotti N,
et al. Rare variants in Toll‑like receptor 7 results in functional impairment
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 12
Biancolellaetal. Human Genomics (2022) 16:19
and downregulation of cytokine‑mediated signaling in COVID‑19
patients. Genes Immun. 2022;23(1):51–6. https:// doi. org/ 10. 1038/
s41435‑ 021‑ 00157‑1 (Epub 2021 Dec 24).
105. Fallerini C, Daga S, Mantovani S, Benetti E, Picchiotti N, Francisci D, et al.
Association of toll‑like receptor 7 variants with life‑threatening COVID‑
19 disease in males: findings from a nested case‑control study. Elife.
2021;2(10):e67569. https:// doi. org/ 10. 7554/ eLife. 67569.
106. Asano T, Boisson B, Onodi F, Matuozzo D, Moncada‑Velez M, Maglorius
Renkilaraj MRL, et al. X‑linked recessive TLR7 deficiency in ~1% of
men under 60 years old with life‑threatening COVID‑19. Sci Immunol.
2021;6(62):eab14348. https:// doi. org/ 10. 1126/ sciim munol. abl43 48.
107. Arkin LM, Moon JJ, Tran JM, Asgari S, O’Farrelly C, Casanova JL, et al.
From your nose to your toes: a review of severe acute respiratory syn‑
drome coronavirus 2 pandemic‑associated pernio. J Invest Dermatol.
2021;141(12):2791–6. https:// doi. org/ 10. 1016/j. jid. 2021. 05. 024 (Epub
2021 Jul 15).
108. Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, et al.
Autoantibodies against type I IFNs in patients with life‑threatening
COVID‑19. Science. 2020;370(6515):eabd4585. https:// doi. org/ 10. 1126/
scien ce. abd45 85.
109. 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/
scien ce. abd45 70.
110. Carapito R, Li R, Helms J, Carapito C, Gujja S, Rolli V, et al. Identification
of driver genes for critical forms of COVID‑19 in a deeply phenotyped
young patient cohort. Sci Transl Med. 2022;14(628):7521. https:// doi.
org/ 10. 1126/ scitr anslm ed. abj75 21.
111. Bastard P, Galerne A, Lefevre‑Utile A, Briand C, Baruchel A, Durand P,
et al. Different clinical presentations and outcomes of disseminated
varicella in children with primary and acquired immunodeficiencies.
Front Immunol. 2020;5(11):595478. https:// doi. org/ 10. 3389/ fimmu.
2020. 595478.
112. Zhou S, Butler‑Laporte G, Nakanishi T, Morrison DR, Afilalo J, Afilalo M,
et al. A Neanderthal OAS1 isoform protects individuals of European
ancestry against COVID‑19 susceptibility and severity. Nat Med.
2021;27(4):659–67. https:// doi. org/ 10. 1038/ s41591‑ 021‑ 01281‑1 (Epub
2021 Feb 25).
113. Magg T, Okano T, Koenig LM, Boehmer DFR, Schwartz SL, Inoue K, et al.
Heterozygous OAS1 gain‑of‑function variants cause an autoinflamma‑
tory immunodeficiency. Sci Immunol. 2021;6(60):eabf9564. https:// doi.
org/ 10. 1126/ sciim munol. abf95 64.
114. Huffman JE, Butler‑Laporte G, Khan A, Pairo‑Castineira E, Drivas TG,
Peloso GM, et al. Multi‑ancestry fine mapping implicates OAS1 splicing
in risk of severe COVID‑19. Nat Genet. 2022;54(2):125–7. https:// doi. org/
10. 1038/ s41588‑ 021‑ 00996‑8 (Epub 2022 Jan 13).
115. Shelton JF, Shastri AJ, Fletez‑Brant K, 23andMe COVID‑19 Team,
Aslibekyan S, Auton A. The UGT2A1/UGT2A2 locus is associated with
COVID‑19‑related loss of smell or taste. Nat Genet. 2022;54(2):121–4.
https:// doi. org/ 10. 1038/ s41588‑ 021‑ 00986‑w.
116. e!Ensembl (2022). Retrieved 19/04, 2022 from https:// www. ensem bl.
org.
117. Kantarcioglu B, Iqbal O, Lewis J, Carter CA, Singh M, Lievano F, et al. An
Update on the status of vaccine development for SARS‑CoV‑2 including
variants. Practical considerations for COVID‑19 special populations. Clin
Appl Thromb Hemost. 2022;28:10760296211056648. https:// doi. org/ 10.
1177/ 10760 29621 10566 48.
118. Barda N, Dagan N, Ben‑Shlomo Y, Kepten E, Waxman J, Ohana R, et al.
Safety of the BNT162b2 mRNA Covid‑19 vaccine in a nationwide
setting. N Engl J Med. 2021;385(12):1078–90. https:// doi. org/ 10. 1056/
NEJMo a2110 475 (Epub 2021 Aug 25).
119. Levine‑Tiefenbrun M, Yelin I, Alapi H, Herzel E, Kuint J, Chodick G,
et al. Waning of SARS‑CoV‑2 booster viral‑load reduction effec‑
tiveness. Nat Commun. 2022;13(1):1237. https:// doi. org/ 10. 1038/
s41467‑ 022‑ 28936‑y.
120. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS‑CoV‑2 and COVID‑
19. Nat Rev Microbiol. 2021;19(3):141–154. https:// doi. org/ 10. 1038/
s41579‑ 020‑ 00459‑7. Epub 2020 Oct 6. Erratum in: Nat Rev Microbiol.
2022 Feb 23.
121. Choi A, Koch M, Wu K, Chu L, Ma L, Hill A, et al. Safety and immuno‑
genicity of SARS‑CoV‑2 variant mRNA vaccine boosters in healthy
adults: an interim analysis. Nat Med. 2021Nov;27(11):2025–31. https://
doi. org/ 10. 1038/ s41591‑ 021‑ 01527‑y (Epub 2021 Sep 15).
122. Barda N, Dagan N, Cohen C, Hernán MA, Lipsitch M, Kohane IS, et al.
Effectiveness of a third dose of the BNT162b2 mRNA COVID‑19 vaccine
for preventing severe outcomes in Israel: an observational study. Lan‑
cet. 2021;398(10316):2093–100. https:// doi. org/ 10. 1016/ S0140‑ 6736(21)
02249‑2 (Epub 2021 Oct 29).
123. Scobie HM, Johnson AG, Suthar AB, Severson R, Alden NB, Balter S, et al.
Monitoring incidence of COVID‑19 Cases, hospitalizations, and deaths,
by vaccination status‑13 U.S. jurisdictions, April 4‑July 17, 2021. MMWR
Morb Mortal Wkly Rep. 2021;70(37):1284–90.
124. Muthukrishnan J, Vardhan V, Mangalesh S, Koley M, Shankar S, Yadav
AK, et al. Vaccination status and COVID‑19 related mortality: a hospital
based cross sectional study. Med J Armed Forces India. 2021;77(Suppl
2):S278–82. https:// doi. org/ 10. 1016/j. mjafi. 2021. 06. 034 (Epub 2021 Jul
26).
125. Klein NP, Stockwell MS, Demarco M, Gaglani M, Kharbanda AB, Irving
SA, et al. Effectiveness of COVID‑19 Pfizer‑BioNTech BNT162b2 mRNA
vaccination in preventing COVID‑19‑associated emergency depart‑
ment and urgent care encounters and hospitalizations among nonim‑
munocompromised children and adolescents aged 5‑17 years ‑ VISION
network, 10 States, April 2021‑January 2022. MMWR Morb Mortal Wkly
Rep. 2022;71(9):352–8.
126. Heudel P, Favier B, Solodky ML, Assaad S, Chaumard N, Tredan O, et al.
Survival and risk of COVID‑19 after SARS‑COV‑2 vaccination in a series
of 2391 cancer patients. Eur J Cancer. 2022;10(165):174–83. https:// doi.
org/ 10. 1016/j. ejca. 2022. 01. 035.
127. Citu IM, Citu C, Gorun F, Sas I, Tomescu L, Neamtu R, et al. Immuno‑
genicity following administration of BNT162b2 and Ad26.COV2.S
COVID‑19 vaccines in the pregnant population during the third trimes‑
ter. Viruses. 2022;14(2):307. https:// doi. org/ 10. 3390/ v1402 0307.
128. Busic N, Lucijanic T, Barsic B, Luksic I, Busic I, Kurdija G, et al. Vaccination
provides protection from respiratory deterioration and death among
hospitalized COVID‑19 patients: Differences between vector and mRNA
vaccines. J Med Virol. 2022. https:// doi. org/ 10. 1002/ jmv. 27666.
129. Ferdinands JM, Rao S, Dixon BE, Mitchell PK, DeSilva MB, Irving SA,
et al. Waning 2‑dose and 3‑dose effectiveness of mRNA vaccines
against COVID‑19‑associated emergency department and urgent care
encounters and hospitalizations among adults during periods of delta
and omicron variant predominance ‑ VISION Network, 10 States, August
2021‑January 2022. MMWR Morb Mortal Wkly Rep. 2022;71(7):255–63.
130. WHO. World Health Organization. Who.int “who.int”.2022; Retrieved
19/04, 2022, from https:// www. who. int/ publi catio ns/m/ item/ draft‑
lands cape‑ of‑ covid‑ 19‑ candi date‑ vacci nes
131. Dragic T, Litwin V, Allaway GP, Mar tin SR, Huang Y, Nagashima KA, et al.
HIV‑1 entry into CD4+ cells is mediated by the chemokine receptor
CC‑CKR‑5. Nature. 1996;381(6584):667–73. https:// doi. org/ 10. 1038/
38166 7a0.
132. Haldane JBS, Jayakar SD. Equilibria under natural selection. J Genet.
1964;59:29–36.
133. Luzzatto L. Genetics of red cells and susceptibility to malaria. Blood.
1979;54(5):961–76.
134. Brenner S, Jacob F, Meselson M. An unstable intermediate carrying
information from genes to ribosomes for protein synthesis. Nature.
1961;13(190):576–81. https:// doi. org/ 10. 1038/ 19057 6a0.
135. Bersani GL. altra epidemia [The parallel epidemic.]. Riv Psichiatr.
2022;57(2):101–5. https:// doi. org/ 10. 1708/ 3790. 37742.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub‑
lished maps and institutional affiliations.
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
... Most of the studied miRNA host-viral interactions with SARS-CoV-2 were based on studies that used bioinformatics rather than experimentation. However, the potential genetic difference between people makes it hard to generalize conclusions about host-viral interactions on a global scale [11,16,22]. ...
Article
Full-text available
Background Mass vaccination and natural immunity reduced the severity of COVID-19 cases. SARS-CoV-2 ongoing genome variations imply the use of confirmatory serologic biomarkers besides PCR for reliable diagnosis. MicroRNA molecules are intrinsic components of the innate immune system. The expression of miR155-5p and miR200c-3p was previously correlated with SARS-CoV-2 pathogenesis. This case-control study was conducted during the third peak of the COVID-19 pandemic in Egypt and aimed to calculate the accuracy of miR155-5p and miR200c-3p as biomarkers for COVID-19. Methods and results Thirty out of 400 COVID-19 patients at a main University hospital in Alexandria were included in the study along with 20 age-matched healthy controls. Plasma samples were collected for total and differential CBC. Relative quantitation of miR155-5p and miR200c-3p expression from WBCs was done by RT-qPCR. The expression of miR155-5p and miR200c-3p was positively correlated and was significantly downregulated in COVID-19 patients compared to the healthy control group (p ˂ 0.005). Both miR155-5p and miR200c-3p were of 76% and 74% accuracy as diagnostic biomarkers of COVID-19, respectively. Regarding the differentiation between mild and moderate cases, their accuracy was 80% and 70%, respectively. Conclusions miR155-5p and miR200c-3p expression can be used to confirm the diagnosis of COVID-19 and discriminate between mild and moderate cases, with a moderate degree of accuracy.
... As of December 2022, the World Health Organization estimates that over 651 million people have been infected with COVID-19, with over 6 million cases of mortality [2]. Recently, however, the pandemic has been well-controlled in some regions through the implementation of public health measures, and in some areas, it is being changed into an endemic [3]. While public health authorities should continue monitoring the virus's spread and implement effective control measures to prevent future outbreaks, those changes mean we also need to pay attention to medical care in patients who have recovered from COVID-19. ...
Article
Full-text available
Background Concern exists about the increasing risk of postoperative pulmonary complications in patients with a history of coronavirus disease 2019 (COVID-19). Objective We conducted a prospective observational study that compared the incidence of postoperative pulmonary complications in patients with and without a history of COVID-19. Methods From August 2022 to November 2022, 244 adult patients undergoing major non-cardiac surgery were enrolled and allocated either to history or no history of COVID-19 groups. For patients without a history of confirming COVID-19 diagnosis, we tested immunoglobulin G to nucleocapsid antigen of SARS-CoV-2 for serology assessment to identify undetected infection. We compared the incidence of postoperative pulmonary complications, defined as a composite of atelectasis, pleural effusion, pulmonary edema, pneumonia, aspiration pneumonitis, and the need for additional oxygen therapy according to a COVID-19 history. Results After excluding 44 patients without a COVID-19 history who were detected as seropositive, 200 patients were finally enrolled in this study, 100 in each group. All subjects with a COVID-19 history experienced no or mild symptoms during infection. The risk of postoperative pulmonary complications was not significantly different between the groups according to the history of COVID-19 (24.0% vs. 26.0%; odds ratio, 0.99; 95% confidence interval, 0.71–1.37; P-value, 0.92). The incidence of postoperative pulmonary complications was also similar (27.3%) in excluded patients owing to being seropositive. Conclusion Our study showed patients with a history of no or mild symptomatic COVID-19 did not show an increased risk of PPCs compared to those without a COVID-19 history. Additional precautions may not be needed to prevent PPCs in those patients.
... The COVID-19 pandemic instigated by the SARS-CoV2 virus remains a public health concern as it transitions to an endemic phase throughout 2023/4. Clinical manifestations can range from pauci-or asymptomatic to an illness necessitating intensive care [1,2]. ...
Article
Full-text available
Introduction: Acute kidney injury (AKI) is a serious and common complication of SARS-CoV-2 infection. Most risk assessment tools for AKI have been developed in the intensive care unit or elderly population. As the COVID-19 pandemic is transitioning into an endemic state, there is an unmet need for prognostic scores tailored to this population. Objectives: Development of a robust predictive model for the occurrence of AKI in hospitalized patients with COVID-19. Patients and methods: Electronic medical records of all adult inpatients admitted between March 2020 and January 2022 were extracted from a large, tertiary care center with reference status in Lesser Poland. We screened 5806 patients with SARS-CoV-2 infection confirmed with polymerase chain reaction test. After excluding subjects with absent serum creatinine values or mild disease course (less than 7 days of inpatient care), 4630 patients were recruited. Data was randomly split into a training (N = 3462) and test (N = 1168) cohort. A random forest model was tuned with feature engineering based on expert advice and metrics evaluated in nested cross-validation to reduce bias. Results: Nested cross-validation yielded an AUC (area under the curve) with a range of 0.793-0.807 and an average performance of 0.798. Model explanation techniques from a global perspective suggest respiratory support, chronic kidney disease and procalcitonin are among the most important variables in permutation tests. Conclusions: The CRACoV-AKI model enables AKI risk stratification among hospitalized patients with COVID-19. Machine learning-based tools may thus offer additional decision-making support for specialist providers.
... First identified in December 2019 [3], the SARS-CoV-2 virus upended our day-to-day activities, initially driving many into isolation, causing interruptions in the provision of essential medical and dental care [4][5][6][7][8][9][10]. As the disease shifts from pandemic to endemic [11], SARS-CoV-2 continues to mutate [12,13]. While initially virulent with high mortality, the virus is less virulent though morbidity and mortality remains high [14,15]. ...
Article
Full-text available
Background During the COVID-19 pandemic, there was a substantial interruption of care, with patients and workers fearful to return to the dental office. As dental practice creates a highly aerosolized environment, the potential for spread of airborne illness is magnified. As a means to increase safety and mitigate risk, pre-visit testing for SARS-CoV-2 has the potential to minimize disease transmission in dental offices. The Pragmatic Return to Effective Dental Infection Control through Testing (PREDICT) Feasibility Study examined the logistics and impact of two different testing mechanisms (laboratory-based PCR viral testing and point-of-care antigen testing) in dental offices. Methods Dental healthcare workers (DHCWs) and patients in four dental offices within the National Dental Practice-based Research Network participated in this prospective study. In addition to electronic surveys, participants in two offices completed POC testing, while participants in two offices used lab-based PCR methods to detect SARS-CoV-2 infection. Analysis was limited to descriptive measures, with median and interquartile ranges reported for Likert scale responses and mean and standard deviation for continuous variables. Results Of the total 72 enrolled, 28 DHCWs and 41 patients completed the protocol. Two patients (4.9%) tested positive prior to their visit, while 2 DHCWs (12.5%) tested positive for SARS-CoV-2 infection at the start of the study. DHCWs and patients shared similar degree of concern (69% and 63%, respectively) for contracting COVID-19 from patients, while patients feared contracting COVID-19 from DHCWs less (49%). Descriptive statistics calculations revealed that saliva, tongue epithelial cells, and nasal swabs were the most desirable specimen collection method; both testing (LAB and POC) protocols took similar amounts of total time to complete; and DHCWs and patients reported feeling more comfortable when both groups were tested. Conclusions While a larger-scale, network study is necessary for generalizability of results, this feasibility study suggests that SARS-CoV-2 testing can be effectively implemented into dental practice workflows and positively impact perception of safety for DHCWs and patients. As new virulent infectious diseases emerge, preparing dental personnel to employ an entire toolbox of risk mitigation strategies, including testing, may have the potential to decrease dental practice closure time, maintaining continuity of dental care services for patients. Trial registration ClinicalTrials.gov: NCT05123742.
... A better understanding of how social work practice has been impacted by the COVID-19 pandemic will enable us to maintain and grow our profession to meet these societal needs within ever-changing practice contexts. Although the acute pandemic phase of has ceded to an endemic phase (Biancolella et al., 2022), literature on how COVID-19 has affected social work practice is still emerging, and there is still much to learn from the lived experiences of social workers. Thus, the purpose of the present quantitative study was twofold: 1) Understand how the COVID-19 pandemic affected the personal and professional lives of social workers practicing in North Carolina; and 2) Determine how the COVID-19 pandemic impacted social worker burnout and organizational and occupational commitment. ...
Article
Full-text available
Since the onset of the COVID-19 pandemic, the demand for social workers in the U.S. and abroad has increased. There is demand for more social workers in North Carolina due to ongoing and increasing mental health, substance use disorder, and child welfare needs. COVID-19 has taken a toll on the personal and professional lives of social workers, and research is needed to understand the pandemic’s effects on burnout and commitment among social workers. The present study sought to understand how the COVID-19 pandemic affected the personal and professional lives of social workers practicing in North Carolina and to determine how the COVID-19 pandemic impacted social worker burnout and organizational and occupational commitment. An online survey was distributed to social workers practicing in North Carolina between February and June of 2022. Social work students recruited 120 eligible participants. Data were analyzed using descriptive statistics, correlations, and multiple regressions. Adjustments to COVID-19 were predictive of work-related burnout and affective commitment when controlling for other factors. Years of practice experience, racial identity, caregiver status, satisfaction with organizational environment, educational attainment, and urbanicity of practice location were also salient predictors across the regression models. North Carolina social workers experienced major adjustments to their personal and professional lives due to the COVID-19 pandemic, which led to negative consequences including increased work-related burnout and less organizational commitment. Additional research – particularly qualitative investigations – is needed to better understand the lived experiences of social workers during the COVID-19 pandemic.
Article
Introduction Even though an MEK inhibitor has been recently launched, neurofibroma still negatively affects the well-being of patients with neurofibromatosis type 1 (NF1). The coronavirus disease 2019 (COVID-19) pandemic resulted in restricted access to medical care. The present study was conducted to investigate the real-world settings of patients with NF1 who underwent surgery with or without restricted medical access during the COVID-19 pandemic. Methods Based on data obtained from medical records, the present study examined 123 and 260 patients who underwent surgery for neurofibromas with and without restricted medical access, respectively. Results The mean numbers of surgeries performed during the periods with and without restricted medical access were 5.8 and 9.8 per month, respectively, and there were 1.18- and 1.46-fold more female patients than male patients for each group, respectively. Regardless of whether medical access was restricted, the majority of patients who underwent surgery were middle-aged females with multiple or severe neurofibromas and mild extracutaneous symptoms. Tumor burden was the most common reason for surgery. However, cutaneous neurofibromas were more likely to be treated than plexiform neurofibromas under restricted medical access. Conclusions Patients with NF1, particularly middle-aged females with severe cutaneous manifestations and mild extracutaneous manifestations, still underwent surgery for neurofibromas regardless of whether medical access was restricted.
Article
Full-text available
Objective To compare vaccination willingness before rollout and 1 year post-rollout uptake among the general population and under-resourced communities in high-income countries. Design A realist review. Data sources Embase, PubMed, Dimensions ai and Google Scholar. Setting High-income countries. Definitions We defined vaccination willingness as the proportion of participants willing or intending to receive vaccines prior to availability. We defined vaccine uptake as the real proportion of the population with complete vaccination as reported by each country until November 2021. Results We included data from 62 studies and 18 high-income countries. For studies conducted among general populations, the proportion of vaccination willingness was 67% (95% CI 62% to 72%). In real-world settings, the overall proportion of vaccine uptake among those countries was 73% (95% CI 69% to 76%). 17 studies reported pre-rollout willingness for under-resourced communities. The summary proportion of vaccination willingness from studies reporting results among people from under-resourced communities was 52% (95% CI 0.46% to 0.57%). Real-world evidence about vaccine uptake after rollout among under-resourced communities was limited. Conclusion Our review emphasises the importance of realist reviews for assessing vaccine acceptance. Limited real-world evidence about vaccine uptake among under-resourced communities in high-income countries is a call to context-specific actions and reporting.
Preprint
Full-text available
Severe-acute-respiratory-syndrome-coronavirus-2 (SARS-CoV-2) is a highly pathogenic and contagious coronavirus that first surfaced in late 2019. The genome encodes four major structural proteins, non-structural proteins and accessory proteins. The nucleocapsid (N) protein of SARS-CoV-2 is an evolutionarily conserved RNA-binding protein that is abundant and plays a critical role in packaging the viral genome. Researchers have explored its potential as a target for therapeutic purposes. People with pre-existing neurological conditions like Parkinson’s disease (PD) and dementia have been recognised as a high-risk population for severe COVID-19 illness as SARS-CoV-2 has been reported to cause deterioration of the symptoms of these diseases. This study aims to identify the shared human interactors of SARS-CoV-2 N protein, PD and dementia. Proteins involved were retrieved from databases, and protein-protein interaction networks were created and visualized in Cytoscape. Individual intersection networks of SARS-CoV-2 N protein with PD and dementia resulted in 46 and 26 proteins, respectively, while intersection networks of SARS-CoV-2 N protein, PD and dementia resulted in 15 common proteins. Seed proteins were identified from network clusters and their Gene Ontology (GO) analysis revealed their involvement in several biological processes. Valosin-containing-protein (VCP) was found to be the only seed protein involved during the co-occurrence of SARS-CoV-2 N protein infection, PD and dementia and is mainly concerned with the regulation of the ubiquitin-proteasome system (UPS). Further, gene enrichment analysis of the identified 15 common proteins was conducted using the DAVID tool, followed by the identification of 7 druggable targets using the Therapeutic Target Database (TTD) and DrugBank. Studying the biological functions of the identified host-protein interactors is crucial for understanding the progression of the disease at a molecular level. Moreover, approved therapeutic compounds against the potential drug target proteins can also be utilized to develop effective treatments.
Article
Full-text available
Here, we provide two methods for monitoring reinfection trends in routine surveillance data to identify signatures of changes in reinfection risk and apply these approaches to data from South Africa’s SARS-CoV-2 epidemic to date. While we found no evidence of increased reinfection risk associated with circulation of Beta (B.1.351) or Delta (B.1.617.2) variants, we find clear, population-level evidence to suggest immune evasion by the Omicron (B.1.1.529) variant in previously infected individuals in South Africa. Reinfections occurring between 01 November 2021 and 31 January 2022 were detected in individuals infected in all three previous waves, and there has been an increase in the risk of having a third infection since mid-November 2021.
Article
Full-text available
There is strong evidence for brain-related abnormalities in COVID-191-13. It remains unknown however whether the impact of SARS-CoV-2 infection can be detected in milder cases, and whether this can reveal possible mechanisms contributing to brain pathology. Here, we investigated brain changes in 785 UK Biobank participants (aged 51-81) imaged twice, including 401 cases who tested positive for infection with SARS-CoV-2 between their two scans, with 141 days on average separating their diagnosis and second scan, and 384 controls. The availability of pre-infection imaging data reduces the likelihood of pre-existing risk factors being misinterpreted as disease effects. We identified significant longitudinal effects when comparing the two groups, including: (i) greater reduction in grey matter thickness and tissue-contrast in the orbitofrontal cortex and parahippocampal gyrus, (ii) greater changes in markers of tissue damage in regions functionally-connected to the primary olfactory cortex, and (iii) greater reduction in global brain size. The infected participants also showed on average larger cognitive decline between the two timepoints. Importantly, these imaging and cognitive longitudinal effects were still seen after excluding the 15 cases who had been hospitalised. These mainly limbic brain imaging results may be the in vivo hallmarks of a degenerative spread of the disease via olfactory pathways, of neuroinflammatory events, or of the loss of sensory input due to anosmia. Whether this deleterious impact can be partially reversed, or whether these effects will persist in the long term, remains to be investigated with additional follow up.
Article
Full-text available
The BNT162b2 COVID-19 vaccine has been shown to reduce viral load of breakthrough infections (BTIs), an important factor affecting infectiousness. This viral-load protective effect has been waning with time post the second vaccine and later restored with a booster shot. It is currently unclear though for how long this regained effectiveness lasts. Analyzing Ct values of SARS-CoV-2 qRT-PCR tests of over 22,000 infections during a Delta-variant-dominant period in Israel, we find that this viral-load reduction effectiveness significantly declines within months post the booster dose. Adjusting for age, sex and calendric date, Ct values of RdRp gene initially increases by 2.7 [CI: 2.3-3.0] relative to unvaccinated in the first month post the booster dose, yet then decays to a difference of 1.3 [CI: 0.7-1.9] in the second month and becomes small and insignificant in the third to fourth months. The rate and magnitude of this post-booster decline in viral-load reduction effectiveness mirror those observed post the second vaccine. These results suggest rapid waning of the booster’s effectiveness in reducing infectiousness, possibly affecting community-level spread of the virus.
Article
Full-text available
Post-acute sequelae of COVID (PASC), usually referred to as 'Long COVID' (a phenotype of COVID-19), is a relatively frequent consequence of SARS-CoV-2 infection, in which symptoms such as breathlessness, fatigue, 'brain fog', tissue damage, inflammation, and coagulopathies (dysfunctions of the blood coagulation system) persist long after the initial infection. It bears similarities to other post-viral syndromes, and to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Many regulatory health bodies still do not recognize this syndrome as a separate disease entity, and refer to it under the broad terminology of 'COVID', although its demographics are quite different from those of acute COVID-19. A few years ago, we discovered that fibrinogen in blood can clot into an anomalous 'amyloid' form of fibrin that (like other β-rich amyloids and prions) is relatively resistant to proteolysis (fibrinolysis). The result, as is strongly manifested in platelet-poor plasma (PPP) of individuals with Long COVID, is extensive fibrin amyloid microclots that can persist, can entrap other proteins, and that may lead to the production of various autoantibodies. These microclots are more-or-less easily measured in PPP with the stain thioflavin T and a simple fluorescence microscope. Although the symptoms of Long COVID are multifarious, we here argue that the ability of these fibrin amyloid microclots (fibrinaloids) to block up capillaries, and thus to limit the passage of red blood cells and hence O2 exchange, can actually underpin the majority of these symptoms. Consistent with this, in a preliminary report, it has been shown that suitable and closely monitored 'triple' anticoagulant therapy that leads to the removal of the microclots also removes the other symptoms. Fibrin amyloid microclots represent a novel and potentially important target for both the understanding and treatment of Long COVID and related disorders.
Article
The paper is a reflection on the psychic phenomenon involving the denial of reality of covid-19 pandemic, the denial of data from scientific research about it and the denial of therapeutic purposes of vaccine and national and international health policies, as it emerges in the frame of the so-called no-vax movements. The possible basic or associated psychopathological pictures are described, analogies and differences respect to classic psychiatric nosology are evaluated, psychological and psychiatric interpretative hypotheses are considered, in what they can to some extent characterize a wide and complex reality, in whose knowledge and management psychiatrists could play a much more relevant role than they actually do.
Article
Background The SARS-CoV-2 Omicron variant became a global concern due to its rapid spread and displacement of the dominant Delta variant. We hypothesized that part of Omicron’s rapid rise was based on its increased ability to cause infections in persons that are vaccinated compared to Delta. Methods We analyzed nasal swab PCR tests for samples collected between 12-26 December 2021 in Connecticut when the proportion of Delta and Omicron variants were relatively equal. We used the spike gene target failure (SGTF) to classify probable Delta and Omicron infections. We fitted an exponential curve to the estimated infections to determine the doubling times for each variant. We compared the test positivity rates for each variant by vaccination status, number of doses, and vaccine manufacturer. Generalized linear models were used to assess factors associated with odds of infection with each variant among persons testing positive for SARS-CoV-2. Findings For infections with high virus copies (Ct < 30) among vaccinated persons, we found higher odds that they were infected with Omicron compared to Delta, and that the odds increased with increased number of vaccine doses. Compared to unvaccinated persons, we found significant reduction in Delta positivity rates after two (43.4-49.1%) and three vaccine doses (81.1%), while we only found a significant reduction in Omicron positivity rates after three doses (62.3%). Conclusion The rapid rise in Omicron infections was likely driven by Omicron’s escape from vaccine-induced immunity. Funding This work was supported by the Centers for Disease Control and Prevention (CDC).
Article
The efficacy of the BNT162b2 (Pfizer-BioNTech) vaccine against laboratory-confirmed COVID-19 exceeded 90% in clinical trials that included children and adolescents aged 5-11, 12-15, and 16-17 years (1-3). Limited real-world data on 2-dose mRNA vaccine effectiveness (VE) in persons aged 12-17 years (referred to as adolescents in this report) have also indicated high levels of protection against SARS-CoV-2 (the virus that causes COVID-19) infection and COVID-19-associated hospitalization (4-6); however, data on VE against the SARS-CoV-2 B.1.1.529 (Omicron) variant and duration of protection are limited. Pfizer-BioNTech VE data are not available for children aged 5-11 years. In partnership with CDC, the VISION Network* examined 39,217 emergency department (ED) and urgent care (UC) encounters and 1,699 hospitalizations† among persons aged 5-17 years with COVID-19-like illness across 10 states during April 9, 2021-January 29, 2022,§ to estimate VE using a case-control test-negative design. Among children aged 5-11 years, VE against laboratory-confirmed COVID-19-associated ED and UC encounters 14-67 days after dose 2 (the longest interval after dose 2 in this age group) was 46%. Among adolescents aged 12-15 and 16-17 years, VE 14-149 days after dose 2 was 83% and 76%, respectively; VE ≥150 days after dose 2 was 38% and 46%, respectively. Among adolescents aged 16-17 years, VE increased to 86% ≥7 days after dose 3 (booster dose). VE against COVID-19-associated ED and UC encounters was substantially lower during the Omicron predominant period than the B.1.617.2 (Delta) predominant period among adolescents aged 12-17 years, with no significant protection ≥150 days after dose 2 during Omicron predominance. However, in adolescents aged 16-17 years, VE during the Omicron predominant period increased to 81% ≥7 days after a third booster dose. During the full study period, including pre-Delta, Delta, and Omicron predominant periods, VE against laboratory-confirmed COVID-19-associated hospitalization among children aged 5-11 years was 74% 14-67 days after dose 2, with wide CIs that included zero. Among adolescents aged 12-15 and 16-17 years, VE 14-149 days after dose 2 was 92% and 94%, respectively; VE ≥150 days after dose 2 was 73% and 88%, respectively. All eligible children and adolescents should remain up to date with recommended COVID-19 vaccinations, including a booster dose for those aged 12-17 years.
Article
Outcomes of 109 hospitalized COVID‐19 patients who received at least one vaccine dose fourteen or more days prior the disease onset were retrospectively compared to control cohort of 109 age, sex and Charlson‐comorbidity‐index matched patients chosen among 2990 total hospitalized patients in our tertiary‐level institution in a period from January to June 2021. Among 109 vaccinated patients, 84 patients were partially and 25 fully vaccinated. Vaccinated patients experienced significantly lower 30‐days mortality (30% vs 49%; hazard ratio (HR) 0.56 (0.37‐0.85); P=0.008), less frequently required high flow oxygen therapy (17% vs 34%; HR 0.45 (0.26‐0.76); P=0.005) and mechanical ventilation (8% vs 18%; HR 0.41 (0.20‐0.88); P=0.027) in comparison to matched cohort of unvaccinated patients. More favorable survival was observed in patients receiving vector in comparison to mRNA vaccine types in unadjusted analysis (30‐days mortality 18% vs 40%; HR 0.45 (0.25‐0.79); P=0.034). In the multivariable Cox regression analysis model both mRNA (HR 0.59 (0.36‐0.98); P=0.041) and vector vaccine types (HR 0.30 (0.15‐0.60); P<0.001) were associated with improved survival in comparison to unvaccinated patients, independently of age (HR 1.03 (1.01‐1.06); P=0.011), male sex (HR 1.78 (1.14‐2.76); P=0.010), severity of illness (HR 2.06 (1.36‐3.10); P<0.001) and functional status on admission (HR 1.42 (1.07‐1.85); P=0.013). This article is protected by copyright. All rights reserved.
Article
Background Headache is a frequent symptoms of coronavirus disease 2019 (COVID-19). Its long-term evolution remains unknown. We aim to evaluate the long-term duration of headache in patients that presented headache during the acute phase of COVID-19. Methods This is a post-hoc multicenter ambisective study including patients from six different third-level hospitals between 1 March and 27 April 2020. Patients completed 9 months of neurological follow-up. Results We included 905 patients. Their median age was 51 (IQR 45–65), 66.5% were female, and 52.7% had a prior history of primary headache. The median duration of headache was 14 (6–39) days; however, the headache persisted after 3 months in 19.0% (95% CI: 16.5–21.8%) and after 9 months in 16.0% (95% confidence interval: 13.7–18.7%). Headache intensity during the acute phase was associated with a more prolonged duration of headache (Hazard ratio 0.655; 95% confidence interval: 0.582–0.737). Conclusion The median duration of headache was 2 weeks, but in approximately a fifth of patients it became persistent and followed a chronic daily pattern.